Next Article in Journal
Oncological Outcomes after Liver Venous Deprivation for Colorectal Liver Metastases: A Single Center Experience
Previous Article in Journal
Fertility Preservation and Long-Term Monitoring of Gonadotoxicity in Girls, Adolescents and Young Adults Undergoing Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation

1
Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
2
CINTESIS—Center for Health Technology and Services Research, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
3
Research Department of the Portuguese League Against Cancer—North (LPCC-NRNorte), Estrada da Circunvalação, 4200-177 Porto, Portugal
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(2), 203; https://doi.org/10.3390/cancers13020203
Submission received: 11 December 2020 / Revised: 31 December 2020 / Accepted: 7 January 2021 / Published: 8 January 2021

Abstract

:

Simple Summary

ASCT2 and LAT1 are amino acid transporters whose impact in cancer has been explored throughout the years. They have been associated with most currently accepted hallmarks of cancer, thus the aim of this review is to report the impact of these transporters in this disease, as well as their clinical significance and applications. ASCT2 and LAT1 have been identified as prognostic factors and potentially as therapeutic targets. In conclusion, the study and development of new inhibitors for these amino acid transporters constitutes a promising approach towards the improvement of cancer treatment and prognosis.

Abstract

The role of the amino acid transporters ASCT2 and LAT1 in cancer has been explored throughout the years. In this review, we report their impact on the hallmarks of cancer, as well as their clinical significance. Overall, both proteins have been associated with cell death resistance through dysregulation of caspases and sustainment of proliferative signaling through mTOR activation. Furthermore, ASCT2 appears to play an important role in cellular energetics regulation, whereas LAT1 expression is associated with angiogenesis and invasion and metastasis activation. The molecular impact of these proteins on the hallmarks of cancer translates into various clinical applications and both transporters have been identified as prognostic factors in many types of cancer. Concerning their role as therapeutic targets, efforts have been undertaken to synthesize competitive or irreversible ASCT2 and LAT1 inhibitors. However, JHP203, a selective inhibitor of the latter, is, to the best of our knowledge, the only compound included in a Phase 1 clinical trial. In conclusion, considering the usefulness of ASCT2 and LAT1 in a variety of cancer-related pathways and cancer therapy/diagnosis, the development and testing of novel inhibitors for these transporters that could be evaluated in clinical trials represents a promising approach to cancer prognosis improvement.

1. Introduction

Protein synthesis is essential to fuel the metabolic needs of cancer cell growth and relies on the maintenance of a homeostatic concentration of cytosolic amino acids, the primary source of cellular nitrogen [1,2,3]. Protein and amino acid synthesis and degradation, as well as amino acid uptake by transporters, are essential homeostasis regulators [1]. The latter are also important in cellular signaling and growth regulation, therefore highlighting potential targets for cancer therapy [3,4].
During the development of cancer and other metabolic diseases, proteins that confer growth and survival advantages, like amino acid transporters, are often overexpressed [3]. These transporters can be found in the plasma membrane or intracellular compartments such as the mitochondria, late endos40omes, lysosomes, and the Golgi apparatus [5]. Their dysregulation affects many functional factors, from intracellular energy metabolism to neurotransmission, and leads to metabolic reprogramming, triggering the carcinogenic process [5]. The neutral amino acid transporters solute carrier family A1 member 5/alanine, serine, cysteine transporter 2 (SLC1A5/ASCT2) and solute carrier family A7 member 5/L-type amino acid transporter 1 (SLC7A5/LAT1) ensure the rapid exchange of different amino acids and the maintenance of an amino acid pool in the cytosol, being likely involved in the development of several malignancies [4,5].
The hallmarks of cancer represent biological capabilities cancer cells acquire during the development of a tumor and help understand the vast diversity of neoplastic diseases [6]. This review will cover the function of ASCT2 and LAT1 transporters, their involvement in cancer, and their association with the hallmarks of cancer. Finally, it will address the clinical significance of both transporters, including their prognostic value and implication in therapy.

1.1. Amino Acid Transport Systems: Focus on Systems ASC and L

In cells, amino acid transport across the plasma membrane is mediated by distinct transport systems [3]. Based on their substrate specificity, regulatory properties, and transport mechanism, amino acid transporters can be classified as sodium (Na+)-dependent neutral amino acid transporters, including systems A, ASC, where ASCT2 is included, B, and N; Na+-independent neutral amino acid transporters, gathering system L, that includes LAT1, and system T; Na+-dependent and -independent anionic amino acid transporters; and Na+-dependent and -independent cationic amino acid transporters [5,7]. Additionally, based on their sequence homology, these transporters can be included in different family groups [3]. Indeed, system ASC proteins belong to the solute carrier family 1 (SLC1) [3]. On the other hand, system L proteins are divided between two families, with LAT1 and LAT2 belonging to the SLC7 family, and LAT3 and LAT4 belonging to the SLC43 family [3].
As previously mentioned, system ASC includes Na+-dependent antiporters, ASCT1 and ASCT2, and it was originally named due to its affinity for the alanine, serine, and cysteine amino acids to differentiate it from system A [3,8]. This sodium ion dependency gives this system access to the energy stores inherent to the gradients of inorganic ions, making it able to generate gradients of amino acids in favor of the cytoplasmic environment [9].
System L activity is attributed to four Na+-independent transporters, LAT1, LAT2, LAT3, and LAT4 [3]. LAT1 and LAT2 have been shown to require the formation of covalent disulfide bridges between their 12 putative membrane-spanning domains and a type-II membrane glycoprotein heavy chain named 4F2hc to form a functional heterodimeric transporter [3]. On the contrary, LAT3 and LAT4 have been shown to not need 4F2hc to be functional [3]. These transporters mediate the uptake of bulky hydrophobic amino acids and are sensible to 2-amino-bicyclo[2.2.1]heptane-2-carboxylic acid (BCH), a synthetic competitive inhibitor [3].

1.1.1. ASCT2: Function and Structure

ASCT2 is encoded by the SLC1A5 gene and its structure shows a homotrimer (Figure 1a,b) [10,11]. It functions as an amino acid exchanger (antiporter) and accepts only neutral amino acids for transport in both directions [12]. Each ASCT2 protomer consists of a transport and a scaffold domain connected by the extracellular loop region 2 (ECL2a and ECL2b) [11]. The former contains the transmembrane segments (TMs) TM3, TM6-TM8 and the helical loops (HP) HP1 and HP2, whereas the latter includes the helices TM1, TM2, TM4, and TM5 [11]. Each transport domain interacts only with the scaffold domain from its own protomer and the ECL2 bridges the two domains, connecting TM3 to TM5 via TM4 (Figure 1c) [11]. There is a different number of Cys residues among SLC1 family members, with the only one conserved corresponding to C363 in ASCT2 [13]. Moreover, the reduction (SH) or oxidation (S-S) of Cys residues allows the switch of the protein state between activated and inactivated [13]. Unlike transporters from other systems, Na+ to K+ or L+ substitution is not tolerated and, contrary to LAT1, it is not inhibited by BCH [8,14]. The mechanism through which this transporter operates remains unclear, nevertheless, it is known that the exchange of amino acids is electroneutral and involves the cotransport of one Na+ ion and one neutral amino acid into the cytosol coupled with the cotransport of also one Na+ ion and one neutral amino acid out of the cell [3].
The ASCT2 transporter shows an asymmetric specificity, with alanine, methionine, and valine being transported only into the intracellular environment and asparagine, glutamate, serine, and threonine being carried in both directions [8]. Cysteine has been shown to work not as a ASCT2 substrate, but as a modulator at slightly higher concentrations, since it triggers efflux by uniport mode [15]. Glutamine is a preferred substrate and it has been shown that its uptake in several cancer cell lines is mediated mainly by this transporter [12].

1.1.2. LAT1: Function and Structure

LAT1 is a heterodimeric amino acid transporter (HAT) that functions as an Na+-independent antiporter [17]. It consists of a functional light chain, LAT1, a 55 kDa protein encoded by the SLC7A5 gene, that links through a conserved disulfide bridge to a heavy chain (4F2hc, also known as CD98), a 68 kDa type II glycoprotein encoded by the SLC3A2 gene, that functions as a chaperone to recruit the functional subunit to the plasma membrane, stabilizing it (Figure 2a) [12,17,18,19,20,21,22,23]. LAT1 consists of 12 TMs, with TM1, TM3, TM6, TM8, and TM10 comprising the inner layer, which is surrounded by TM2, TM4, TM5, TM7, TM9, TM11, and TM12, the outer layer [24]. On the other hand, 4F2hc only comprises a single TM [24]. The complex also contains several loop and helical segments (H1–H4) [25]. The disulfide bridge forms between two conserved cysteine residues, one in the loop between transmembrane helices 3 and 4 of LAT1 and the other in 4F2hc (Figure 2b) [17]. LAT1 belongs to the SLC7 gene family, is not glycosylated, and exhibits an intracellular N- and C-terminus, whereas the latter belongs to the SLC3 gene family, is N-glycosylated, and presents an intracellular N-terminal and a bulky extracellular C-terminal [8,12]. Its substate-binding site consists of a proximal pocket that accommodates primary side chains, a distal pocket that functions as a binding site for hydrophobic secondary substitutions, and the positive and negative poles of two short helices that recognize the carboxyl and amino groups of the substrates [23]. LAT1 has a high affinity for bulky branched-chain and aromatic amino acids, particularly leucine, phenylalanine, and tryptophan, and can transport both D- and L-amino acid enantiomers [3,12,22,26]. Alanine, proline, and charged amino acids are not recognized by this transporter and it has lower affinity for glutamine and threonine [8]. As an obligatory amino acid exchanger with a 1:1 stoichiometry and lower affinity for substrates located within the cell its velocity is dependent on the concentration of the intracellular amino acids [3]. LAT1 has been shown to transport several drugs across the blood-brain barrier (BBB), being expressed at 100-fold higher levels in BBB endothelial cells than in other tissues [22].

1.2. ASCT2 and LAT1: Their Expression in Cancer

In a review by Fuchs et al. [3], ASCT2, LAT1, and sodium-coupled amino acid transporter 5 (SNAT5) expression stood out as significantly enhanced in tumors compared to their normal counterparts. However, whereas the latter had not been verified in other studies, ASCT2 and LAT1 expression pattern was similar in a variety of cancerous tissues (Table 1) [3].
Comparing to LAT1, ASCT2 transports a much wider range of substrates, namely glutamine as previously mentioned [3]. In fact, increased expression of this transporter has been documented in tissues where glutamine plays a particularly important role in metabolism, such as the liver, brain and the epithelial cells of the gut [3]. Moreover, enhanced ASCT2 expression has also been reported in cancers derived from tissues where this protein is not usually present [14].
Upregulation of both LAT1 and 4F2hc has been reported in cancer tissues or proliferative cells [12,27]. LAT1 has been detected in RERF-LC-MA lung small cell carcinoma cells, leukemia cell lines, T24 bladder carcinoma cells, and HeLa uterine cervical carcinoma cells and its expression has been correlated with the size of metastatic distant tumors in rats, thus being suggested as a potential therapeutic target for many cancers [3].

2. ASCT2 and LAT1: Their Contribution to the Hallmarks of Cancer

The hallmarks of cancer have been proposed by Hanahan and Weinberg and consist of biological concepts that allow the characterization of the heterogeneous and complex nature of cancer [6]. They consist of six capabilities that promote tumor progression (resisting cell death, sustaining proliferative signaling, activating invasion and metastasis, inducing angiogenesis, enabling replicative immortality, and evading growth suppressors) and are accompanied by two enabling characteristics (tumor-promoting inflammation and genomic instability and mutation) and two emerging hallmarks (avoiding immune destruction and dysregulating cellular energetics) that represent the important aspect of the constitution and signaling of the tumor microenvironment [6,66].
The role of the amino acid transporters ASCT2 and LAT1 has not yet been explored in the context of two of the six original hallmarks, enabling replicative immortality and evading growth suppressors, nor of the enabling characteristics. In this section, we will explore the association between ASCT2 and LAT1 and these distinctive and complementary hallmarks of cancer (Figure 3).

2.1. Resisting Cell Death

Evasion of programmed cell death or apoptosis represents a major way of increasing the rate of cell proliferation during tumor progression [66]. Overall, apoptosis can result from an intrinsic or extrinsic pathway and both culminate in the activation of proteases named caspases [109]. The former, also known as the mitochondrial pathway, is mainly regulated by the B-cell lymphoma 2 (BCL-2) family of proteins, which can be activated by a variety of cellular stress signals, such as hypoxia, DNA damage and nutrient deprivation, and includes the anti-apoptotic proteins BCL-2 and BCL-xL and the pro-apoptotic effectors BCL-2-associated X protein (BAX) and BCL-2 antagonist/killer (BAK), among others [109]. The extracellular pathway, on the other hand, can result from the binding of specific ligands to death receptors from the tumor necrosis factor (TNF) superfamily located in the cell surface, activating them [109]. Regarding the caspases involved in programmed cell death, caspase-2, -8, -9, and -10 are considered apoptosis initiators, whereas caspase-3, -6, and -7 are apoptosis executioners or effectors [110].
ASCT2 knockdown has shown to play a role in the mitochondrial pathway of apoptosis, as it has been associated with decreased BCL-2 levels, increased BAX expression and a loss of matrix metallopeptidases (MMPs), which results from the mitochondrial membrane permeabilization facilitated by BAX [79]. Wang et al. [79], using a reactive oxygen species (ROS) scavenger, NAC, reported a reversion of the effects of ASCT2 knockdown in gastric cancer cell lines, supporting that this protein ablation leads to oxidative stress and contributes to apoptosis. That involvement was also observed in colorectal cancer cell lines using a synthetic small interfering RNA (siRNA) to attenuate the expression of SLC1A5 [29] and in non-small cell lung cancer (NSCLC) using the l-γ-glutamyl-p-nitroanilide (GPNA) ASCT2 inhibitor [80]. Topotecan (TPT) is a DNA topoisomerase I inhibitor that has shown anti-cancer effects on gastric cancer and the results from its testing suggest that ASCT2 is an anti-cancer target for this compound, as it presented the same effects as ASCT2 knockdown [79]. Furthermore, the silencing of this protein with antisense RNA in hepatic adenocarcinoma cells did not appear to be attributed to glutamine or overall amino acid deprivation, thus suggesting a crucial role of ASCT2 in cell apoptosis beyond maintaining of amino acid homeostasis [81]. In fact, whereas both ASCT2 silencing and glutamine starvation promoted caspases-2 and -3 activity, there was no evidence for caspases-8 and -9 activities in glutamine-starved cells [82]. Moreover, both caspases-2 and -9 are upregulated in response to ASCT2 suppression and showed to be involved in the intrinsic pathway of apoptosis in response to oxidative stress, which in turn may result from the loss of glutathione (GSH), an antioxidant, due to the decrease of its precursors glutamine and cysteine, amino acids transported by ASCT2 (Figure 4) [82]. Knockdown or inhibition of this transporter has been associated with increased levels of ROS and decreased levels of GSH in gastrointestinal cell lines and with significant loss of mitochondrial potential in lung cancer cells [79,80,83]. The epidermal growth factor receptor (EGFR) is involved in downstream signal transduction of several pathways upon binding of a specific ligand, which may result in cell proliferation or evasion of apoptosis [84]. ASCT2 has been identified as an EGFR-associated protein that can be co-targeted by cetuximab, an EGFR antibody approved for metastatic human head and neck squamous cell carcinoma treatment, sensitizing the cancer cells to ROS-induced apoptosis through reduction of intracellular levels of glutamine and, consequently, of glutathione [85].
Dysregulation of LAT1 transporter has also been shown to affect caspase activity and, consequently, apoptosis [20]. Treatment of the KB human oral epidermoid carcinoma cells, Saos2 human osteogenic sarcoma cells and C6 rat glioma cells with BCH induced DNA fragmentation and increased TUNEL-positive cells [86]. Moreover, LAT1 inhibition led to the activation through proteolytic cleavage of caspase-3 and -7, which are synthesized as inactive proenzymes, in KB and C6 cells, inducing apoptotic cell death [86]. In Saos2 cells, this induction only depended on caspase-7 activation [86]. Similarly, BCH treatment also induced TUNEL-positive apoptotic cells from a human malignant glioma cell line [87]. Furthermore, this effect correlated with cleavage induction of both caspase-3 and its intrinsic substrate PARP [87].

2.2. Sustaining Proliferative Signaling

The ability to sustain chronic proliferation depends on the release of growth-promoting signals and cell cycle progression [6]. The phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin complex (mTORC) 1 pathway is critical for cell growth, integrating signals from a variety of factors, such as energy status and growth factors, and is highly supported by nutrients such as amino acids, particularly leucine, which is required for the activation of mTORC1 and is transported into the cell by LAT1 [3,111]. mTORC1 and mTORC2, the two known mTOR complexes, are inhibited during amino acid starvation, leading to a decrease in mRNA and protein biosynthesis resulting from alterations in the phosphorylation pattern of their downstream effectors, namely ribosomal protein S6 kinase beta-1 (S6K1) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) [5]. mTORC1 activation occurs at the lysosomal membrane and its rate-limiting step is the uptake of l-glutamine via ASCT2 [5,112]. After 1–2 min, that amino acid leaves the cell, leading to the uptake of l-leucine by LAT1 and the rapid activation of S6K1 through mTORC1 [5].
The role of ASCT2 in tumor growth, namely its association with the inhibition of mTOR pathway, has been observed in breast cancer [113], melanoma [114], hepatoma [81], endometrial carcinoma [31], acute myeloid leukemia [115], gastric cancer [116], and prostate cancer [38], through either lentiviral transduction of a short hairpin RNA (shRNA) against ASCT2 or treatment with ASCT2 inhibitors (benzylserine or GPNA, for example). In fact, a study by Avissar et al. [88] showed that the PI3K pathway induced glutamine transport and ASCT2 expression in the human enterocytes cell surface. C-Myc is a well-characterized oncogene also involved in tumor cell growth [89]. It is a transcriptional factor frequently overexpressed in cancer and its levels have been correlated with ASCT2 and mTORC1 activation [89]. Additionally, the tumor suppressor retinoblastoma protein (Rb) is involved in cell cycle arrest by disabling the E2F family of cell cycle-promoting transcription factors and an inverse association with ASCT2 overexpression has been reported [90]. Furthermore, upregulation of SNAT1 and SNAT2, members of the transport System A and of the SLC38 family, was reported in ASCT2-deficient cells [91,92]. The substrate specificity of these transporters overlaps that of ASCT2 and they appear to explain the normal mTOR signaling observed in some cells lacking ASCT2 [91].
Leucine is a regulator of mTORC1, as previously mentioned, and also a major substrate for LAT1, which mediates its uptake in exchange for glutamine [5]. Pharmacological inhibition of LAT1, using the selective inhibitor JPH203, significantly reduced cell proliferation associated with mTORC1 pathway in six human cell lines without reducing CD98 expression [93]. In fact, Milkereit et al. [94] showed that LAT1-4F2hc is recruited to lysosomes by the lysosomal-associated transmembrane protein 4b (LAPTM4b), promoting leucine uptake into those organelles and mTORC1 activation. LAT1 inhibition has been associated with reduced cellular proliferation, often associated with reduced phosphorylation level of mTOR and its downstream effectors in several cancer models, including lung [52,95], colorectal [43] and breast cancer [96]. Similarly, LAT1 expression has been associated with increased tumor size and tumor cell growth rates [87,97]. On the other hand, Fan et al. [117] observed little impact of BCH treatment on ovarian cancer cells proliferation. Besides LAT1, this inhibitor is also able to block the activity of other leucine-accepting transporters and the authors conclude that LAT1-mediated amino acid transport alone might have limited impact on anchorage-dependent cancer cell proliferation in tumors [117]. Concerning other pathways, LAT1 inhibition by the same compound induced cell cycle arrest at G1 phase in oral cancer cells by inhibiting the cyclin D2-cyclin-dependent protein kinase 6 (CDK6) complex and increasing p27 expression, a CDK inhibitor [98]. The cyclin/CDK complexes play major roles in cell cycle events, as the binding between those molecules constitutes a target of checkpoint pathways that ensure cell cycle progression [99]. EGFR mutations have been found in lung cancer and they have been shown to have significant responses to EGFR tyrosine kinase inhibitors (TKIs), such as gebitinib and erlotinib [84]. Imai et al. [95] observed higher LAT1 expression in NSCLC samples without EGFR mutations, which might be associated with TKIs refractoriness and a poor prognosis in combination of LAT1 expression and EGFR wild-type.
Since LAT1 is an obligatory exchanger and that leucine uptake is dependent on the intracellular concentration of the exchange substrates, functional amino acid transporter-coupling between ASCT2 and LAT1 has been proposed [118]. In that model, ASCT2 drives glutamine uptake, which serves as an exchange substrate for the LAT1-mediated leucine transport to the intracellular environment, allowing the activation of the mTORC1 pathway and cellular growth, as previously mentioned [118]. A reciprocal regulatory connection has also been observed between ASCT2, LAT1 and mTOR in prostate cancer, hepatoma, lymphoma and leukemic cells, since glutamine starvation-induced LAT1-mediated transport of amino acids or expression and/or leucine deprivation induced the same effects on ASCT2 [3,38,67]. However, Cormerais et al. [119] did not observe a disruption in LAT1 or mTORC1 activity nor in leucine uptake following ASCT2 knockout in colon LS174T and lung A549 adenocarcinoma cells. Nevertheless, ASCT2 ablation decreased tumor growth, suggesting a LAT1-independent role in tumor proliferation [119]. Overall, despite their dominant role in amino acid transport and involvement in cell proliferation, ASCT2 and LAT1 have been classified as “harmonizers” instead of drivers of amino acid signaling and accumulation, since cells seem to adapt to the knockout of those transporters [120].

2.3. Activating Invasion and Metastasis

Local invasion and distant metastasis reflect the tumor progression to higher grades of malignancy, representing the major cause of morbidity and death in cancer patients, and usually represent alterations in cell-cell adhesion and the attachment to the extracellular matrix (ECM) [6].
There are few published studies reporting the association between ASCT2 and invasion and metastasis and the underlying mechanism remains elusive. Nevertheless, a higher expression of SLC1A5 in gastric cancer tissues has been correlated with clinicopathological features such as local invasion and lymph node metastasis [33]. Furthermore, SLC1A5 downregulation in vitro resulted in significant inhibition of gastric cells invasion and migration [33].
Similarly, LAT1 expression has been found to be significantly higher in metastatic sites compared to primary tumors in a variety of cancers [41,58,111,121]. In parallel, higher LAT1 expression has been associated with an increase in metastatic lesion diameter [122]. Blockade of this transporter efficiently inhibited anchorage-independent growth of ovarian cancer cell lines, suggesting an important role in proliferation under non-adhesive conditions as the ones involved in migration and metastasis [117]. Moreover, cholangiocarcinoma cells ability to migrate and invade was reduced by a downregulation of LAT1 expression through shRNA and it is suggested that it may involve 4F2hc-related signaling, since this molecule interacts with β1 integrin and forms a complex that takes part in Akt signaling activation, contributing to carcinogenesis [73]. Cells lacking LAT1 showed a reduction in cell migration due to a decrease in phosphorylation and consequent activation of ERK1/2 signaling pathway, regulated by the reorganization of the collagen matrix that is mediated by the α1β1 integrin [73]. Overexpressed LAT1 together with 4F2hc has been reported as necessary for metastasis in colon cancer patients [74]. Ding et al. [75] determined that the SLC7A5 transcript stability was increased by genetic suppressor element 1 (GSE1) in a post-transcriptional manner independent from direct binding. GSE1 is positively associated with many clinicopathological features, namely lymph node metastasis, clinical stage, depth of invasion and histological grade and LAT1 appears to mediate its functions and to be positively regulated by GSE1 [75]. Wang et al. [47] identified LAT1 as a downstream target of the CRK-like (CRKL) protein, a promoter of gastric cancer. Depletion of CRKL in a gastric cancer cell line resulted in impairment of SLC7A5 expression and suppression of cell motility [47].

2.4. Inducing Angiogenesis

Tumor cells with high proliferation rates require high levels of nutrients and oxygen, as well as an efficient ability to dispose of carbon dioxide and metabolic wastes [6]. These needs are addressed by the process of angiogenesis, which allows neovascularization associated with the tumor [6].
The expression level of both ASCT2 and LAT1 showed a significant correlation with CD147, an inductor of matrix metalloproteinases and tumor angiogenesis [76]. LAT1 expression correlated with that of vascular endothelial growth factor (VEGF), a marker of angiogenesis, in thymic carcinomas [77], colon [74] and lung cancers [63]. Furthermore, LAT1 has been detected in vascular endothelial and its expression level has been associated with glioma microvessel density [78].

2.5. Avoiding Immune Destruction

Immune surveillance recognizes and eliminates the majority of incipient cancer cells through the constant monitoring of tissues and cells by the immune system [6]. However, developing tumors appear to be able to evade immune recognition and thus avoid destruction [6]. In fact, an increase in tumor incidence has been observed in patients with deficiency of natural killer (NK) cells, CD8+ cytotoxic T lymphocytes (CTLs) and Th1 cells [6].
Interestingly, both ASCT2 and LAT1 have been reported as mediators of amino acid supply to activated T cells and important factors for T cell differentiation into Th1 and Th17 cells [7,68]. Moreover, ASCT2 transporter is required for mTORC1 signaling activation stimulated by T-cell receptor (TCR) and CD28, but not in other T cell activation pathways, making it particularly important in naïve T cells although dispensable in effector T cells, where it might be functionally redundant with other amino acid transporters [69]. LAT1 expression has been shown to be induced by CD3, which forms a complex with the TCR and allows its signaling, and CD28, a heterodimer that not only binds to integrin-β and mediates survival and growth through adhesive signals but is also involved in amino acid transport [68,70,71]. T cells need to achieve high levels of amino acid incorporation for normal immune responses, therefore requiring LAT1 to maintain their activated state [68]. In fact, T cells appear to use LAT1 to sustain the high demand for nutrients required for immune reactions in the same way cancer cells also prefer using this transporter to efficiently proliferate [68]. Despite ASCT2 importance in T cell activation, some studies have shown that, in ASCT2-knockout mice, this amino acid transporter is not required for T and B cell development, B cell proliferation and antibody production, suggesting that the immune system of cancer patients might tolerate the inhibition of this transporter [67,72].

2.6. Dysregulating Cellular Energetics

The uncontrollable proliferation rates that characterize cancer cells require adjustments in cell energy metabolism so that it can fuel the growth [6]. Glutamine is a non-essential amino acid that cells can synthesize de novo [123]. Nevertheless, it is one of the most important amino acids in the human body, being a source of both reduced nitrogen for biosynthetic reactions and of carbon for the mitochondrial tricarboxylic acid (TCA) cycle, also serving as a precursor to nucleotide and lipid synthesis [8,123,124]. Glutamine catabolism involves the conversion of glutamine to glutamate, a precursor of the TCA cycle intermediate α-ketoglutarate (α-KG), by cytosolic glutamine aminotransferases or mitochondrial glutaminases [123]. On the other hand, α-KG can be used to endogenously synthesize glutamine via the enzymes glutamate dehydrogenase and glutamine synthetase [125]. The need for glutamine is enhanced when cells undergo a high proliferation rate, with many tumors depending on glutaminolysis to fuel their energetic and metabolic necessities [5,14]. This process yields increased levels of downstream metabolites such as α-KG, pyruvate and lactate [5]. Additionally, the TCA cycle, where glutamine ends its fate, generates NADH and FADH2 that provide electrons for mitochondrial ATP generation [5,125]. Simultaneously, glutaminolysis has been considered a hallmark of cancer and its increase in cancer cells has been associated with an upregulation of some amino acid transporters, particularly ASCT2, whose targeting may lead to promising anti-cancer treatments [5,14].
Alteration of glutamine uptake has been reported as a result of the action of proto-oncogenes or tumor suppressors, similar to their action in stimulating cell proliferation as aforementioned [126]. C-Myc has been found to bind to promoter elements of the gene encoding ASCT2, resulting in higher levels of SLC1A5 mRNA and enhanced glutamine uptake [100]. On the other hand, deletion of the tumor suppressors belonging to the Rb family alters glutamine metabolism by increasing the uptake of this amino acid through E2F transcription factor 3 (E2F3) binding to the SLC1A5 promoter [90]. RNF5 is an E3 ubiquitin ligase associated with the endoplasmic reticulum involved in the regulation of protein stability and clearance, allowing the functioning of various cellular processes [101]. This protein has been found to play a role in ASCT2 degradation, also resulting in reduced glutamine uptake, induced by protein misfolding in response to endoplasmic reticulum stress [101]. There are also published studies on the influence of the tumor microenvironment on glutamine metabolism, contributing to tumor progression, involving cytokines and chemokines [102,127]. Interleukins (ILs) are secreted proteins that interact with specific receptors and allow communication between cells [128]. IL-4 is a Th2 cytokine that interacts with the IL-4 receptor (IL-4R) and has been reported as a regulator of high ASCT2 mRNA and protein expression, enhancing glutamine uptake in breast cancer cells [102]. IL-3, required for survival and proliferation of hematopoietic progenitor cells, binds to IL-3Rα, a glucose-dependent receptor, and signaling through this receptor induces ASCT2 [103]. Moreover, Jak inhibition repressed glutamine uptake, completely blocking ASCT2 induction, suggesting that IL-3 action occurs via the Jak/signal transducer and activator of transcription (STAT) pathway [103]. Ren et al. [104] have shown that neuroblastomas with amplification of the MYCN gene, which is associated with high levels of the N-Myc protein, rely on high amounts of glutamine to sustain cell viability, the TCA cycle and biosynthetic activities and that those high amounts are supported by ASCT2 activation. The activation of this transporter appeared to occur via activating transcription factor 4 (ATF4), a member of the cyclic AMP response element binding protein (CREB) family frequently induced in response to hypoxia, endoplasmic reticulum (ER) and oxidative stress and nutrient deprivation, in coordination with N-Myc [104]. There are also studies reporting the epigenetic modulation of tumor glutamine metabolism via ASCT2. In a study by Dong et al. [105], inhibition of the tumor suppressor microRNA-137 (miR-137) through methylation of its promoter by DNA methyltransferases (DNMTs) together with methyl-CpG-binding protein 2 (MeCP2) led to ASCT2 upregulation and, consequently, glutamine metabolism reactivation, identifying miR-137 as a regulator of glutaminolysis by targeting ASCT2 [105]. Interestingly, this miR has also been identified as a regulator of ferroptosis, an iron-dependent form of cell death genetically and morphologically different from apoptosis, by directly targeting ASCT2 in melanoma cells [106]. miR-137 suppressed ferroptosis, inhibiting the ability of melanoma cells to colonize in vitro and develop tumors in a mouse model [106]. Ferroptosis inhibition led to ASCT2 overexpression, which rescued the suppressing effect of miR-137 in this form of cell death [106].
Despite its high affinity for glutamine and the fact that SLC1A5 knockout did decrease the uptake of this amino acid, blockade of ASCT2 in leukemic cells resulted in a much more pronounced effect on global cell metabolism which cannot be explained solely by glutamine transport [67]. Other amino acids play important roles in biosynthesis and intracellular redox homeostasis in cancer cells, such as cysteine, isoleucine, threonine and valine, and they have been found to significantly decrease in SLC1A5-knockout cells [67]. Furthermore, the pentose phosphate pathway, the glycolytic pathway, as well as the methylation cycle, had decreased rates in leukemic cells in the same study by Ni et al. [67].
Leucine plays important signaling roles, promoting protein synthesis through the activation of the mTOR pathway, enhancing mitochondrial biogenesis and augmenting fatty acid oxidation [129]. As previously mentioned, this amino acid can be transported into the cell by LAT1 [129]. Treatment of NSCLC cells with delta-tocotrienol (δT) inhibited both LAT1 and ASCT2 expression, resulting in a significant decrease in leucine concentration [107]. Furthermore, treatment of breast cancer cells with JPH203, a tyrosine analog and selective LAT1 inhibitor, limited the amount of leucine, and also tyrosine, that could maintain protein production or enter the TCA cycle, proving to be beneficial in combination with other mTOR inhibitors and/or endocrine therapies for breast cancer treatment [108].

3. ASCT2 and LAT1: Their Clinical Significance in Cancer

After acknowledging the potential role of the neutral amino acid transporters addressed here on the hallmarks of cancer, the focus of researchers and pharmaceuticals on ASCT2 and LAT1 clinical relevance is to be expected [130] (Figure 5). If these proteins are overexpressed in tumor cells, their blockade would likely lead to amino acid deprivation, impairing protein synthesis and, consequently, growth, while not affecting the biology of normal cells [131]. Moreover, drugs based on transporters are proposed to reduce possible side effects and enhance clinical efficacy [132].

3.1. ASCT2 and LAT1 as Prognostic Biomarkers in Cancer

The prognostic value of LAT1 in cancer has recently been the subject of two meta-analyses [133,134]. Those reviews reported associations between high LAT1 expression and poor overall, cancer-specific, disease-free and progression-free survival, as well as clinicopathological features such as stage, tumor size, lymphatic and vascular invasion, tumor differentiation, Ki-67, 4F2hc, CD34, p53 and ASCT2 [133,134]. LAT1, besides hepatocellular [39,48], lung [49,50,51,60,62,63,64,135,136], colorectal [44,137,138], renal [42,59], ovarian [40,139], breast [57], tongue [140] and pancreatic cancer [54,55], has also been identified as a prognostic marker for multiple myeloma [141], melanoma [142], cholangiocarcinoma [143], biliary tract [144,145], laryngeal [146], bladder [147], prostate [65,148,149], thymic [150] cancer and endometrioid carcinoma [46], even though the latter showed contradicting results.
Although with fewer published studies, ASCT2 has been identified as a prognostic factor in hepatocellular [34], lung [35], colorectal [151], gastric [33], renal [28], ovarian [36,152], breast [153], tongue [140] and pancreatic cancer [76]. Furthermore, genetic variants in the SLC1A5 gene have also been implicated in hepatocellular carcinoma prognosis, with stage I patients carrying the rs2070246 TT genotype showing higher overall survival (OS) than carriers of CC genotype [154]. Concerning the latter, contradicting results had been previously reported by Watanabe et al. [45], who suggested a low significance of LAT1 as a prognostic marker since its expression had been low in poorly differentiated adenocarcinomas and correlated with neither proliferative activity nor the International Federation of Gynecology and Obstetrics stage.
Furthermore, co-expression of both transporters, ASCT2 and LAT1, has also been identified as an independent prognostic factor for lung adenocarcinoma [155] and surgically resected esophageal squamous cell carcinoma [156]. In fact, the combined expression of the transporters presented an overall poorer prognosis compared to the single-positive expression of ASCT2 or LAT1 in both models [155,156].

3.2. LAT1 as a Predictive Factor in Cancer Treatment

The relationship between the expression level of amino acid transporters, particularly of LAT1, and response to chemotherapy in cancer patients as been reported [53]. In NSCLC, LAT1 expression was significantly associated with resistance to platinum-based chemotherapy in patients with postoperative recurrence [53]. Higher expression LAT1 has also been identified as an independent factor for chemoresistance in ovarian clear cell carcinomas [139]. A similar role in chemotherapy resistance has been reported for LAT1 in patients with metastatic or recurrent pancreatic ductal adenocarcinoma after surgical resection, with all patients with high LAT1 expression being identified as non-responders to fluorouracil (5-FU) and gemcitabine [149].

3.3. ASCT2 and LAT1 Therapeutic Targets in Cancer

The research on the ASCT2 and LAT1 transporters as novel therapeutic targets for cancer therapy has been reported in the last five years (Table 2) [132,157,158,159]. The drug design of targets for these transporters usually follows an approach based on substrate analogues, which act as competitive inhibitors [159]. Several amino acid analogues have been reported to competitively bind to the ASCT2 substrate-binding site, impairing the amino acid transport, such as benzylserine [160], GPNA, the most potent compound of a series of glutamine analogues synthesized by Esslinger et al. [161], and V-9302, which selectively targets ASCT2 and represents the only inhibitor of this transporter that reached a preclinical phase [83,159], as well as other molecules based on sulfonamide/sulfonic acid ester scaffolds [162]. The major limitations of this type of inhibitors is the fact that (1) endogenous substrates can displace them from the binding pocket, impeding their effect; and (2) these transporters usually recognize more than one amino acid as substrate [159]. Thus, it is important to design compounds that are able to irreversibly target and block ASCT2 transport. l-phenylglycine and its analogues have been identified as non-substrate molecules that selectively and effectively inhibit the function of this transporter [163]. Furthermore, analogues of the amino acid proline, which is not a substrate for ASCT2, such as benzylproline-derived compounds have also been found as potential inhibitors [164,165]. Due to the strong reactivity of ASCT2 toward metals like mercury and its derivatives that form bonds with cysteine residues, the design of inhibitors that exploit this interaction could also play an important role in the irreversible inhibition of this transporter [14]. The use of monoclonal antibodies (MAbs) has also shown potential as an effective treatment for some cancers, namely in the suppression of glutamine-dependent growth of colorectal cancer cells (KM4008, KM4012 and KM4018) [166] and ASCT2-expressing gastric cancer (KM8094) [56,167].
Efforts have also been undertaken to synthesize potential LAT1 inhibitors due to the well-documented overexpression of this transporter in cancer [168]. Interestingly, GPNA, the widely used ASCT2 inhibitor, as already mentioned, has also been shown to impair the uptake of essential amino acids through other transporters, namely LAT1, lowering the cell content of these nutrients and potentially affecting mTORC1 activity [168]. Moreover, δT also inhibits both ASCT2 and LAT1 by impairing glutamine uptake, resulting in the induction of apoptosis and inhibition of cell proliferation due to mTOR pathway dysregulation [107]. BCH is a nonmetabolizable leucine analogue that has been shown to diminish growth in a variety of cancer cells [132,157,169]. Nevertheless, the concentration required to suppress cancer cell growth is extremely high and it constitutes a rather unspecific inhibitor of all the L-type amino acid transporters (LAT1-4) [157,158]. The anti-cancer potential of KYT-0353 or JHP203, a highly selective LAT1 inhibitor produced through synthetic chemistry and in vitro screening based on triiodothyronine (T3), has been reported in osteosarcoma [170], leukemic [171], oral cancer [172], gastric cancer [173], and colon cancer [173,174] cells, among others [157]. The thyroid hormone derivative 3-iodo-l-tyrosine has been identified as a potent LAT1 inhibitor through cis-inhibition and cell-proliferation experiments [175]. Other compounds have also been reported in recent years, including other inhibitors based on the T3 structure [176], with highlight to SKN103, and based on dithiazole and dithiazine compounds [177]. To the best of our knowledge, only the LAT1 inhibitor JPH203 is being tested in clinical trial setting [178]. In the in-human Phase 1 clinical trial study published by Okano et al. [178], clinical trial registration UMIN000016546, JPH203 was not only reported to have promising activity against biliary tract cancer, but was also well-tolerated in these patients. This compound is predominantly acetylated by N-acetyltransferase 2, which may exhibit distinct phenotypes according to its speed (rapid, intermediate, or slow) before it is excreted in bile [178]. Those phenotypes might be useful to predict JPH203 safety and efficacy [178].

3.4. LAT1 in Cancer Diagnosis and Drug Delivery

The potential of LAT1 for enhanced delivery of nanoparticles into the cancer cells for diagnostic and therapeutic purposes has been previously addressed [157,186,187]. Phenylalanine-coupled solid lipid nanoparticles (SLN) loaded with doxorubicin, a chemotherapy drug, have been shown to target LAT1 in the blood-brain barrier, as well as in brain cancer cells, where it is highly expressed, significantly enhancing the delivery of that drug and its anti-cancer activity [188]. The potential of this transporter as a nanoparticle target, either directed for chemotherapy or photothermal therapy via multi-branched gold nanoparticles, has also been reported for breast cancer treatment [189,190]. Furthermore, the uptake of liposomes modified with a LAT1-targeting polymer with thermoresponsive properties has also been enhanced in HeLa cells, suggesting the usefulness of this transporter as a target for drug delivery [191].
Interestingly, LAT1 has also the potential to be useful in cancer diagnosis due to its ability to transport radio-labeled (18F- or 11C-) amino acid analogues that function as positron emission tomography (PET) probes, allowing their visualization inside the body [158]. 2-18F-fluoro-2-deoxy-D-glucose ([18F]FDG) was one of the most commonly used PET probes, based on the fact that tumor cells consume high amounts of glucose, which led to false-positive results, especially in organs with higher uptake of glucose, like the brain [158]. To overcome this drawback, novel probes, including the amino acid analogues L-4-borono-2-18F-fluorophenylalanine (18F-FBPA) [192], (S)-2-amino-3-[3-(2-18F-fluoroethoxy)-4-iodophenyl]-2-methylpropanoic acid (18F-FIMP) [193] and 2-[18F]-2-fluoroethyl-L-phenylalanine (2-[18F]FELP), attracted attention [194].

4. Conclusions

The importance of amino acid transporters in cancer has been explored in the last two decades. However, there is a limited number of studies supporting the involvement of ASCT2 and LAT1 in tumor development for each cancer model. These transporters have been associated with a variety of hallmarks of cancer, such as resisting cell death, sustaining proliferative signaling, activating invasion and metastasis, inducing angiogenesis, avoiding immune destruction and dysregulating cellular energetics. Moreover, the clinical significance of these transporters has been explored in a variety of cancers. Due to their usefulness for cancer therapy and/or diagnosis, as well as their association with chemotherapy response, efforts should be undertaken to develop and test inhibitors that can be evaluated in clinical trials, such as the LAT1 inhibitor JHP203, hopefully enabling the improvement of cancer prognosis.

Funding

This work was financed by the European Regional Development Fund (ERDF) through the Northern Region Operational Program (NORTE2020) (NORTE-01-0247-FEDER-033399). Furthermore, Carina Pereira is a research fellowship holder (SFRH/BPD/114803/2016) supported by Fundação para a Ciência e Tecnologia (FCT), co-financed by European Social Funds (ESF) and national funds of MCTES under the Human Strategic Reference Framework (POCH).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

4E-BP1 Eukaryotic translation initiation factor 4E-binding protein 1
5-FUFluorouracil
α-KGα-Ketoglutarate
δTδ-Tocotrienol
ASCT2Alanine, serine, cysteine transporter 2
ATF4Activating transcription factor 4
BAKBCL-2 antagonist/killer
BAXBCL-2-associated X protein
BBBBlood-brain barrier
BCH2-Aminobicyclo [2,2,1]heptane-2-carboxylic acid
BCL-2B-cell lymphoma 2
CDK6Cyclin-dependent protein kinase 6
CREBCyclic AMP response element binding protein
CRKLCRK-like
CTLCytotoxic T lymphocytes
DNMTDeoxyribonucleic acid methyltransferase
E2F3E2F transcription factor 3
ECMExtracellular matrix
EGFREpidermal growth factor receptor
EREndoplasmic reticulum
GPNAL-γ-glutamyl-p-nitroanilide
GSE1Genetic suppressor element 1
GSHGlutathione
HATHeterodimeric amino acid transporter
ILInterleukin
IL-RInterleukin receptor
LAPTM4bLysosomal-associated transmembrane protein 4b
LAT1L-type amino acid transporter 1
MAbMonoclonal antibody
MeCP2Methyl-cpg-binding protein 2
miRMicro ribonucleic acid
MMPMatrix metallopeptidase
mTORCMammalian target of rapamycin complex
NKNatural killer
NSCLCNon-small cell lung cancer
OSOverall survival
PETPositron emission tomography
PI-3KPhosphoinositide 3-kinase
RbRetinoblastoma protein
ROSReactive oxygen species
S6K1Ribosomal protein S6 kinase beta-1
shRNAShort hairpin ribonucleic acid
siRNASmall interfering ribonucleic acid
SLCSolute carrier family
SLC1A5Solute carrier family A1 member 5
SLC7A5Solute carrier family A7 member 5
SLNSolid lipid nanoparticles
SNATSodium-coupled amino acid transporter
STATSignal transducer and activator of transcription
TCATricarboxylic acid
TCRT-cell receptor
TKITyrosine kinase inhibitor
TMTransmembrane segment
TNFTumor necrosis factor
TPTTopotecan
VEGFVascular endothelial growth factor

References

  1. Bröer, S.; Bröer, A. Amino acid homeostasis and signalling in mammalian cells and organisms. Biochem. J. 2017, 474, 1935–1963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hosios, A.M.; Hecht, V.C.; Danai, L.V.; Johnson, M.O.; Rathmell, J.C.; Steinhauser, M.L.; Manalis, S.R.; Vander Heiden, M.G. Amino acids rather than glucose account for the majority of cell mass in proliferating mammalian cells. Dev. Cell 2016, 36, 540–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fuchs, B.C.; Bode, B.P. Amino acid transporters ASCT2 and LAT1 in cancer: Partners in crime? Semin. Cancer Biol. 2005, 15, 254–266. [Google Scholar] [CrossRef] [PubMed]
  4. Bröer, A.; Gauthier-Coles, G.; Rahimi, F.; van Geldermalsen, M.; Dorsch, D.; Wegener, A.; Holst, J.; Bröer, S. Ablation of the ASCT2 (SLC1A5) gene encoding a neutral amino acid transporter reveals transporter plasticity and redundancy in cancer cells. J. Biol. Chem. 2019, 294, 4012–4026. [Google Scholar] [CrossRef] [Green Version]
  5. Kandasamy, P.; Gyimesi, G.; Kanai, Y.; Hediger, M.A. Amino acid transporters revisited: New views in health and disease. Trends Biochem. Sci. 2018, 43, 752–789. [Google Scholar] [CrossRef]
  6. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  7. Ren, W.; Liu, G.; Yin, J.; Tan, B.; Wu, G.; Bazer, F.W.; Peng, Y.; Yin, Y. Amino-acid transporters in T-cell activation and differentiation. Cell Death Dis. 2017, 8, e2655. [Google Scholar] [CrossRef]
  8. Leke, R.; Schousboe, A. The glutamine transporters and their role in the glutamate/GABA-glutamine Cycle. Adv. Neurobiol. 2016, 13, 223–257. [Google Scholar] [CrossRef]
  9. Christensen, H.N. Role of amino acid transport and countertransport in nutrition and metabolism. Physiol. Rev. 1990, 70, 43–77. [Google Scholar] [CrossRef]
  10. Liu, Y.; Zhao, T.; Li, Z.; Wang, L.; Yuan, S.; Sun, L. The role of ASCT2 in cancer: A review. Eur. J. Pharmacol. 2018, 837, 81–87. [Google Scholar] [CrossRef]
  11. Yu, X.; Plotnikova, O.; Bonin, P.D.; Subashi, T.A.; McLellan, T.J.; Dumlao, D.; Che, Y.; Dong, Y.Y.; Carpenter, E.P.; West, G.M.; et al. Cryo-EM structures of the human glutamine transporter SLC1A5 (ASCT2) in the outward-facing conformation. eLife 2019, 8, e48120. [Google Scholar] [CrossRef] [PubMed]
  12. Ganapathy, V.; Thangaraju, M.; Prasad, P.D. Nutrient transporters in cancer: Relevance to Warburg hypothesis and beyond. Pharmacol. Ther. 2009, 121, 29–40. [Google Scholar] [CrossRef] [PubMed]
  13. Scalise, M.; Pochini, L.; Console, L.; Pappacoda, G.; Pingitore, P.; Hedfalk, K.; Indiveri, C. Cys site-directed mutagenesis of the human SLC1A5 (ASCT2) transporter: Structure/function relationships and crucial role of Cys467 for redox sensing and glutamine transport. Int. J. Mol. Sci. 2018, 19, 648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Scalise, M.; Pochini, L.; Console, L.; Losso, M.A.; Indiveri, C. The human SLC1A5 (ASCT2) amino acid transporter: From function to structure and role in cell biology. Front. Cell Dev. Biol. 2018, 6. [Google Scholar] [CrossRef] [PubMed]
  15. Scalise, M.; Pochini, L.; Pingitore, P.; Hedfalk, K.; Indiveri, C. Cysteine is not a substrate but a specific modulator of human ASCT2 (SLC1A5) transporter. FEBS Lett. 2015, 589, 3617–3623. [Google Scholar] [CrossRef]
  16. Garaeva, A.A.; Oostergetel, G.T.; Gati, C.; Guskov, A.; Paulino, C.; Slotboom, D.J. Cryo-EM structure of the human neutral amino acid transporter ASCT2. Nat. Struct. Mol. Biol. 2018, 25, 515–521. [Google Scholar] [CrossRef]
  17. Chiduza, G.N.; Johnson, R.M.; Wright, G.S.A.; Antonyuk, S.V.; Muench, S.P.; Hasnain, S.S. LAT1 (SLC7A5) and CD98hc (SLC3A2) complex dynamics revealed by single-particle cryo-EM. Acta Crystallogr. D Struct. Biol. 2019, 75, 660–669. [Google Scholar] [CrossRef]
  18. Napolitano, L.; Scalise, M.; Galluccio, M.; Pochini, L.; Albanese, L.M.; Indiveri, C. LAT1 is the transport competent unit of the LAT1/CD98 heterodimeric amino acid transporter. Int. J. Biochem. Cell Biol. 2015, 67, 25–33. [Google Scholar] [CrossRef]
  19. Yanagida, O.; Kanai, Y.; Chairoungdua, A.; Kim, D.K.; Segawa, H.; Nii, T.; Cha, S.H.; Matsuo, H.; Fukushima, J.I.; Fukasawa, Y.; et al. Human L-type amino acid transporter 1 (LAT1): Characterization of function and expression in tumor cell lines. Biochim. Biophys. Acta Biomembr. 2001, 1514, 291–302. [Google Scholar] [CrossRef] [Green Version]
  20. Zhao, Y.; Wang, L.; Pan, J. The role of L-type amino acid transporter 1 in human tumors. Intractable Rare Dis. Res. 2015, 4, 165–169. [Google Scholar] [CrossRef] [Green Version]
  21. Kanai, Y.; Segawa, H.; Miyamoto, K.I.; Uchino, H.; Takeda, E.; Endou, H. Expression Cloning and Characterization of a Transporter for Large Neutral Amino Acids Activated by the Heavy Chain of 4F2 Antigen (CD98). J. Biol. Chem. 1998, 273, 23629–23632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Chien, H.C.; Colas, C.; Finke, K.; Springer, S.; Stoner, L.; Zur, A.A.; Venteicher, B.; Campbell, J.; Hall, C.; Flint, A.; et al. Reevaluating the Substrate Specificity of the L-Type Amino Acid Transporter (LAT1). J. Med. Chem. 2018, 61, 7358–7373. [Google Scholar] [CrossRef] [PubMed]
  23. Lee, Y.; Wiriyasermkul, P.; Jin, C.; Quan, L.; Ohgaki, R.; Okuda, S.; Kusakizako, T.; Nishizawa, T.; Oda, K.; Ishitani, R.; et al. Cryo-EM structure of the human L-type amino acid transporter 1 in complex with glycoprotein CD98hc. Nat. Struct. Mol. Biol. 2019, 26, 510–517. [Google Scholar] [CrossRef] [PubMed]
  24. Singh, N.; Ecker, G.F. Insights into the structure, function, and ligand discovery of the large neutral amino acid transporter 1, LAT1. Int. J. Mol. Sci. 2018, 19, 1278. [Google Scholar] [CrossRef] [Green Version]
  25. Yan, R.; Zhao, X.; Lei, J.; Zhou, Q. Structure of the human LAT1–4F2hc heteromeric amino acid transporter complex. Nature 2019, 568, 127–130. [Google Scholar] [CrossRef]
  26. Scalise, M.; Galluccio, M.; Console, L.; Pochini, L.; Indiveri, C. The human SLC7A5 (LAT1): The intriguing histidine/large neutral amino acid transporter and its relevance to human health. Front. Chem. 2018, 6. [Google Scholar] [CrossRef]
  27. Ichinoe, M.; Mikami, T.; Yoshida, T.; Igawa, I.; Tsuruta, T.; Nakada, N.; Anzai, N.; Suzuki, Y.; Endou, H.; Okayasu, I. High expression of L-type amino-acid transporter 1 (LAT1) in gastric carcinomas: Comparison with non-cancerous lesions. Pathol. Int. 2011, 61, 281–289. [Google Scholar] [CrossRef]
  28. Liu, Y.; Yang, L.; An, H.; Chang, Y.; Zhang, W.; Zhu, Y.; Xu, L.; Xu, J. High expression of Solute Carrier Family 1, member 5 (SLC1A5) is associated with poor prognosis in clear-cell renal cell carcinoma. Sci. Rep. 2015, 5, 16954. [Google Scholar] [CrossRef] [Green Version]
  29. Huang, F.; Zhao, Y.; Zhao, J.; Wu, S.; Jiang, Y.; Ma, H.; Zhang, T. Upregulated SLC1A5 promotes cell growth and survival in colorectal cancer. Int. J. Clin. Exp. Pathol. 2014, 7, 6006–6014. [Google Scholar]
  30. Schulte, M.L.; Hight, M.R.; Ayers, G.D.; Liu, Q.; Shyr, Y.; Washington, M.K.; Manning, H.C. Non-invasive glutamine PET reflects pharmacological inhibition of BRAF(V600E) in vivo. Mol. Imaging Biol. 2017, 19, 421–428. [Google Scholar] [CrossRef]
  31. Marshall, A.D.; van Geldermalsen, M.; Otte, N.J.; Lum, T.; Vellozzi, M.; Thoeng, A.; Pang, A.; Nagarajah, R.; Zhang, B.; Wang, Q.; et al. ASCT2 regulates glutamine uptake and cell growth in endometrial carcinoma. Oncogenesis 2017, 6, e367. [Google Scholar] [CrossRef] [PubMed]
  32. Risinger, J.; Allard, J.; Chandran, U.; Day, R.; Chandramouli, G.; Miller, C.; Zahn, C.; Oliver, J.; Litzi, T.; Marcus, C.; et al. Gene expression analysis of early stage endometrial cancers reveals unique transcripts associated with grade and histology but not depth of invasion. Front. Oncol. 2013, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lu, J.; Chen, M.; Tao, Z.; Gao, S.; Li, Y.; Cao, Y.; Lu, C.; Zou, X. Effects of targeting SLC1A5 on inhibiting gastric cancer growth and tumor development in vitro and in vivo. Oncotarget 2017, 8, 76458–76467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sun, H.W.; Yu, X.J.; Wu, W.C.; Chen, J.; Shi, M.; Zheng, L.; Xu, J. GLUT1 and ASCT2 as predictors for prognosis of hepatocellular carcinoma. PLoS ONE 2016, 11, e0168907. [Google Scholar] [CrossRef] [PubMed]
  35. Shimizu, K.; Kaira, K.; Tomizawa, Y.; Sunaga, N.; Kawashima, O.; Oriuchi, N.; Tominaga, H.; Nagamori, S.; Kanai, Y.; Yamada, M.; et al. ASC amino-acid transporter 2 (ASCT2) as a novel prognostic marker in non-small cell lung cancer. Br. J. Cancer 2014, 110, 2030–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Guo, H.; Xu, Y.; Wang, F.; Shen, Z.; Tuo, X.; Qian, H.; Wang, H.; Wang, K. Clinical associations between ASCT2 and p-mTOR in the pathogenesis and prognosis of epithelial ovarian cancer. Oncol. Rep. 2018, 40, 3725–3733. [Google Scholar] [CrossRef] [Green Version]
  37. Kaira, K.; Sunose, Y.; Arakawa, K.; Sunaga, N.; Shimizu, K.; Tominaga, H.; Oriuchi, N.; Nagamori, S.; Kanai, Y.; Oyama, T.; et al. Clinicopathological significance of ASC amino acid transporter-2 expression in pancreatic ductal carcinoma. Histopathology 2015, 66, 234–243. [Google Scholar] [CrossRef]
  38. Wang, Q.; Hardie, R.A.; Hoy, A.J.; van Geldermalsen, M.; Gao, D.; Fazli, L.; Sadowski, M.C.; Balaban, S.; Schreuder, M.; Nagarajah, R.; et al. Targeting ASCT2-mediated glutamine uptake blocks prostate cancer growth and tumour development. J. Pathol. 2015, 236, 278–289. [Google Scholar] [CrossRef] [Green Version]
  39. Namikawa, M.; Kakizaki, S.; Kaira, K.; Tojima, H.; Yamazaki, Y.; Horiguchi, N.; Sato, K.; Oriuchi, N.; Tominaga, H.; Sunose, Y.; et al. Expression of amino acid transporters (LAT1, ASCT2 and xCT) as clinical significance in hepatocellular carcinoma. Hepatol. Res. 2015, 45, 1014–1022. [Google Scholar] [CrossRef]
  40. Kaira, K.; Nakamura, K.; Hirakawa, T.; Imai, H.; Tominaga, H.; Oriuchi, N.; Nagamori, S.; Kanai, Y.; Tsukamoto, N.; Oyama, T.; et al. Prognostic significance of L-type amino acid transporter 1 (LAT1) expression in patients with ovarian tumors. Am. J. Transl. Res. 2015, 7, 1161–1171. [Google Scholar]
  41. Papin-Michault, C.; Bonnetaud, C.; Dufour, M.; Almairac, F.; Coutts, M.; Patouraux, S.; Virolle, T.; Darcourt, J.; Burel-Vandenbos, F. Study of LAT1 expression in brain metastases: Towards a better understanding of the results of positron emission tomography using amino acid tracers. PLoS ONE 2016, 11, e0157139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Betsunoh, H.; Fukuda, T.; Anzai, N.; Nishihara, D.; Mizuno, T.; Yuki, H.; Masuda, A.; Yamaguchi, Y.; Abe, H.; Yashi, M.; et al. Increased expression of system large amino acid transporter (LAT)-1 mRNA is associated with invasive potential and unfavorable prognosis of human clear cell renal cell carcinoma. BMC Cancer 2013, 13, 509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Hayase, S.; Kumamoto, K.; Saito, K.; Kofunato, Y.; Sato, Y.; Okayama, H.; Miyamoto, K.; Ohki, S.; Takenoshita, S. L-type amino acid transporter 1 expression is upregulated and associated with cellular proliferation in colorectal cancer. Oncol. Lett. 2017, 14, 7410–7416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Sakata, T.; Hana, K.; Mikami, T.; Yoshida, T.; Endou, H.; Okayasu, I. Positive correlation of expression of L-type amino-acid transporter 1 with colorectal tumor progression and prognosis: Higher expression in sporadic colorectal tumors compared with ulcerative colitis-associated neoplasia. Pathol. Res. Pract. 2020, 216, 152972. [Google Scholar] [CrossRef] [PubMed]
  45. Watanabe, J.; Yokoyama, Y.; Futagami, M.; Mizunuma, H.; Yoshioka, H.; Washiya, K.; Hana, K.; Endou, H.; Okayasu, I. L-type amino acid transporter 1 expression increases in well-differentiated but decreases in poorly differentiated endometrial endometrioid adenocarcinoma and shows an inverse correlation with p53 expression. Int. J. Gynecol. Cancer 2014, 24, 659–663. [Google Scholar] [CrossRef] [PubMed]
  46. Sato, K.; Miyamoto, M.; Takano, M.; Tsuda, H. Correlation of high LAT1 expression with the prognosis of endometrioid carcinoma of the uterine corpus. Virchows Arch. 2020, 477, 421–427. [Google Scholar] [CrossRef]
  47. Wang, J.; Fei, X.; Wu, W.; Chen, X.; Su, L.; Zhu, Z.; Zhou, Y. SLC7A5 functions as a downstream target modulated by CRKL in metastasis process of gastric cancer SGC-7901 Cells. PLoS ONE 2016, 11, e0166147. [Google Scholar] [CrossRef]
  48. Li, J.; Qiang, J.; Chen, S.F.; Wang, X.; Fu, J.; Chen, Y. The impact of L-type amino acid transporter 1 (LAT1) in human hepatocellular carcinoma. Tumor Biol. 2013, 34, 2977–2981. [Google Scholar] [CrossRef]
  49. Kaira, K.; Oriuchi, N.; Imai, H.; Shimizu, K.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Tanaka, S.; Ishizuka, T.; Kanai, Y.; et al. Prognostic significance of L-type amino acid transporter 1 expression in resectable stage I–III nonsmall cell lung cancer. Br. J. Cancer 2008, 98, 742–748. [Google Scholar] [CrossRef]
  50. Imai, H.; Kaira, K.; Oriuchi, N.; Yanagitani, N.; Sunaga, N.; Ishizuka, T.; Kanai, Y.; Endou, H.; Nakajima, T.; Mori, M. L-type amino acid transporter 1 expression is a prognostic marker in patients with surgically resected stage I non-small cell lung cancer. Histopathology 2009, 54, 804–813. [Google Scholar] [CrossRef]
  51. Takeuchi, K.; Ogata, S.; Nakanishi, K.; Ozeki, Y.; Hiroi, S.; Tominaga, S.; Aida, S.; Matsuo, H.; Sakata, T.; Kawai, T. LAT1 expression in non-small-cell lung carcinomas: Analyses by semiquantitative reverse transcription-PCR (237 cases) and immunohistochemistry (295 cases). Lung Cancer 2010, 68, 58–65. [Google Scholar] [CrossRef] [PubMed]
  52. Kaira, K.; Oriuchi, N.; Takahashi, T.; Nakagawa, K.; Ohde, Y.; Okumura, T.; Murakami, H.; Shukuya, T.; Kenmotsu, H.; Naito, T.; et al. LAT1 expression is closely associated with hypoxic markers and mTOR in resected non-small cell lung cancer. Am. J. Transl. Res. 2011, 3, 468–478. [Google Scholar] [PubMed]
  53. Kaira, K.; Takahashi, T.; Murakami, H.; Shukuya, T.; Kenmotsu, H.; Naito, T.; Oriuchi, N.; Kanai, Y.; Endo, M.; Kondo, H.; et al. Relationship between LAT1 expression and response to platinum-based chemotherapy in non-small cell lung cancer patients with postoperative recurrence. Anticancer Res. 2011, 31, 3775–3782. [Google Scholar]
  54. Kaira, K.; Sunose, Y.; Arakawa, K.; Ogawa, T.; Sunaga, N.; Shimizu, K.; Tominaga, H.; Oriuchi, N.; Itoh, H.; Nagamori, S.; et al. Prognostic significance of L-type amino-acid transporter 1 expression in surgically resected pancreatic cancer. Br. J. Cancer 2012, 107, 632–638. [Google Scholar] [CrossRef] [PubMed]
  55. Yanagisawa, N.; Ichinoe, M.; Mikami, T.; Nakada, N.; Hana, K.; Koizumi, W.; Endou, H.; Okayasu, I. High expression of L-type amino acid transporter 1 (LAT1) predicts poor prognosis in pancreatic ductal adenocarcinomas. J. Clin. Pathol. 2012, 65, 1019–1023. [Google Scholar] [CrossRef]
  56. Osanai-Sasakawa, A.; Hosomi, K.; Sumitomo, Y.; Takizawa, T.; Tomura-Suruki, S.; Imaizumi, M.; Kasai, N.; Poh, T.W.; Yamano, K.; Yong, W.P.; et al. An anti-ASCT2 monoclonal antibody suppresses gastric cancer growth by inducing oxidative stress and antibody dependent cellular toxicity in preclinical models. Am. J. Cancer Res. 2018, 8, 1499–1513. [Google Scholar]
  57. Furuya, M.; Horiguchi, J.; Nakajima, H.; Kanai, Y.; Oyama, T. Correlation of L-type amino acid transporter 1 and CD98 expression with triple negative breast cancer prognosis. Cancer Sci. 2012, 103, 382–389. [Google Scholar] [CrossRef]
  58. Kaji, M.; Kabir-Salmani, M.; Anzai, N.; Jin, C.J.; Akimoto, Y.; Horita, A.; Sakamoto, A.; Kanai, Y.; Sakurai, H.; Iwashita, M. Properties of L-type amino acid transporter 1 in epidermal ovarian cancer. Int. J. Gynecol. Cancer 2010, 20, 329–336. [Google Scholar] [CrossRef]
  59. Higuchi, K.; Sakamoto, S.; Ando, K.; Maimaiti, M.; Takeshita, N.; Okunushi, K.; Reien, Y.; Imamura, Y.; Sazuka, T.; Nakamura, K.; et al. Characterization of the expression of LAT1 as a prognostic indicator and a therapeutic target in renal cell carcinoma. Sci. Rep. 2019, 9, 16776. [Google Scholar] [CrossRef]
  60. Kaira, K.; Oriuchi, N.; Imai, H.; Shimizu, K.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Kawashima, O.; Iijima, H.; Ishizuka, T.; et al. Expression of L-type amino acid transporter 1 (LAT1) in neuroendocrine tumors of the lung. Pathol. Res. Pract. 2008, 204, 553–561. [Google Scholar] [CrossRef]
  61. Kaira, K.; Oriuchi, N.; Imai, H.; Shimizu, K.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Kawashima, O.; Kamide, Y.; Ishizuka, T.; et al. CD98 expression is associated with poor prognosis in resected non-small-cell lung cancer with lymph node metastases. Ann. Surg. Oncol. 2009, 16, 3473–3481. [Google Scholar] [CrossRef] [PubMed]
  62. Kaira, K.; Oriuchi, N.; Imai, H.; Shimizu, K.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Ishizuka, T.; Kanai, Y.; Endou, H.; et al. Prognostic significance of L-type amino acid transporter 1 (LAT1) and 4F2 heavy chain (CD98) expression in early stage squamous cell carcinoma of the lung. Cancer Sci. 2009, 100, 249–254. [Google Scholar] [CrossRef] [PubMed]
  63. Kaira, K.; Oriuchi, N.; Imai, H.; Shimizu, K.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Kawashima, O.; Kamide, Y.; Ishizuka, T.; et al. Prognostic significance of L-type amino acid transporter 1 (LAT1) and 4F2 heavy chain (CD98) expression in surgically resectable stage III non-small cell lung cancer. Exp. Ther. Med. 2010, 1, 799–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kaira, K.; Oriuchi, N.; Shimizu, K.; Imai, H.; Tominaga, H.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Ishizuka, T.; Kanai, Y.; et al. Comparison of L-type amino acid transporter 1 expression and L-[3-18F]-α-methyl tyrosine uptake in outcome of non-small cell lung cancer. Nucl. Med. Biol. 2010, 37, 911–916. [Google Scholar] [CrossRef]
  65. Sakata, T.; Ferdous, G.; Tsuruta, T.; Satoh, T.; Baba, S.; Muto, T.; Ueno, A.; Kanai, Y.; Endou, H.; Okayasu, I. L-type amino-acid transporter 1 as a novel biomarker for high-grade malignancy in prostate cancer. Pathol. Int. 2009, 59, 7–18. [Google Scholar] [CrossRef]
  66. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
  67. Ni, F.; Yu, W.M.; Li, Z.; Graham, D.K.; Jin, L.; Kang, S.; Rossi, M.R.; Li, S.; Broxmeyer, H.E.; Qu, C.K. Critical role of ASCT2-mediated amino acid metabolism in promoting leukaemia development and progression. Nat. Metab. 2019, 1, 390–403. [Google Scholar] [CrossRef]
  68. Hayashi, K.; Jutabha, P.; Endou, H.; Sagara, H.; Anzai, N. LAT1 is a critical transporter of essential amino acids for immune reactions in activated human T cells. J. Immunol. 2013, 191, 4080–4085. [Google Scholar] [CrossRef]
  69. Nakaya, M.; Xiao, Y.; Zhou, X.; Chang, J.H.; Chang, M.; Cheng, X.; Blonska, M.; Lin, X.; Sun, S.C. Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. Immunity 2014, 40, 692–705. [Google Scholar] [CrossRef] [Green Version]
  70. Van der Merwe, P.A.; Dushek, O. Mechanisms for T cell receptor triggering. Nat. Rev. Immunol. 2011, 11, 47–55. [Google Scholar] [CrossRef]
  71. Cantor, J.M.; Ginsberg, M.H. CD98 at the crossroads of adaptive immunity and cancer. J. Cell Sci. 2012, 125, 1373–1382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Masle-Farquhar, E.; Bröer, A.; Yabas, M.; Enders, A.; Bröer, S. ASCT2 (SLC1A5)-deficient mice have normal B-cell development, proliferation, and antibody production. Front. Immunol. 2017, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Janpipatkul, K.; Suksen, K.; Borwornpinyo, S.; Jearawiriyapaisarn, N.; Hongeng, S.; Piyachaturawat, P.; Chairoungdua, A. Downregulation of LAT1 expression suppresses cholangiocarcinoma cell invasion and migration. Cell. Signal. 2014, 26, 1668–1679. [Google Scholar] [CrossRef] [PubMed]
  74. Kaira, K.; Oriuchi, N.; Imai, H.; Shimizu, K.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Tanaka, S.; Ishizuka, T.; Kanai, Y.; et al. L-type amino acid transporter 1 and CD98 expression in primary and metastatic sites of human neoplasms. Cancer Sci. 2008, 99, 2380–2386. [Google Scholar] [CrossRef] [PubMed]
  75. Ding, K.; Tan, S.; Huang, X.; Wang, X.; Li, X.; Fan, R.; Zhu, Y.; Lobie, P.E.; Wang, W.; Wu, Z. GSE1 predicts poor survival outcome in gastric cancer patients by SLC7A5 enhancement of tumor growth and metastasis. J. Biol. Chem. 2018, 293, 3949–3964. [Google Scholar] [CrossRef] [Green Version]
  76. Kaira, K.; Arakawa, K.; Shimizu, K.; Oriuchi, N.; Nagamori, S.; Kanai, Y.; Oyama, T.; Takeyoshi, I. Relationship between CD147 and expression of amino acid transporters (LAT1 and ASCT2) in patients with pancreatic cancer. Am. J. Transl. Res. 2015, 7, 356–363. [Google Scholar]
  77. Kaira, K.; Oriuchi, N.; Imai, H.; Shimizu, K.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Ishizuka, T.; Kanai, Y.; Endou, H.; et al. L-type amino acid transporter 1 (LAT1) is frequently expressed in thymic carcinomas but is absent in thymomas. J. Surg. Oncol. 2009, 99, 433–438. [Google Scholar] [CrossRef]
  78. Haining, Z.; Kawai, N.; Miyake, K.; Okada, M.; Okubo, S.; Zhang, X.; Fei, Z.; Tamiya, T. Relation of LAT1/4F2hc expression with pathological grade, proliferation and angiogenesis in human gliomas. BMC Clin. Pathol. 2012, 12, 4. [Google Scholar] [CrossRef] [Green Version]
  79. Wang, L.; Liu, Y.; Zhao, T.L.; Li, Z.Z.; He, J.Y.; Zhang, B.J.; Du, H.Z.; Jiang, J.W.; Yuan, S.T.; Sun, L. Topotecan induces apoptosis via ASCT2 mediated oxidative stress in gastric cancer. Phytomedicine 2019, 57, 117–128. [Google Scholar] [CrossRef]
  80. Hassanein, M.; Qian, J.; Hoeksema, M.D.; Wang, J.; Jacobovitz, M.; Ji, X.; Harris, F.T.; Harris, B.K.; Boyd, K.L.; Chen, H.; et al. Targeting SLC1a5-mediated glutamine dependence in non-small cell lung cancer. Int. J. Cancer 2015, 137, 1587–1597. [Google Scholar] [CrossRef] [Green Version]
  81. Fuchs, B.C.; Finger, R.E.; Onan, M.C.; Bode, B.P. ASCT2 silencing regulates mammalian target-of-rapamycin growth and survival signaling in human hepatoma cells. Am. J. Physiol. Cell Physiol. 2007, 293, C55–C63. [Google Scholar] [CrossRef] [PubMed]
  82. Fuchs, B.C.; Perez, J.C.; Suetterlin, J.E.; Chaudhry, S.B.; Bode, B.P. Inducible antisense RNA targeting amino acid transporter ATB0/ASCT2 elicits apoptosis in human hepatoma cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 286, G467–G478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Schulte, M.L.; Fu, A.; Zhao, P.; Li, J.; Geng, L.; Smith, S.T.; Kondo, J.; Coffey, R.J.; Johnson, M.O.; Rathmell, J.C.; et al. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat. Med. 2018, 24, 194–202. [Google Scholar] [CrossRef] [PubMed]
  84. Bethune, G.; Bethune, D.; Ridgway, N.; Xu, Z. Epidermal growth factor receptor (EGFR) in lung cancer: An overview and update. J. Thorac. Dis. 2010, 2, 48–51. [Google Scholar] [PubMed]
  85. Lu, H.; Li, X.; Lu, Y.; Qiu, S.; Fan, Z. ASCT2 (SLC1A5) is an EGFR-associated protein that can be co-targeted by cetuximab to sensitize cancer cells to ROS-induced apoptosis. Cancer Lett. 2016, 381, 23–30. [Google Scholar] [CrossRef] [Green Version]
  86. Kim, C.S.; Cho, S.H.; Chun, H.S.; Lee, S.Y.; Endou, H.; Kanai, Y.; Kim, D.K. BCH, an inhibitor of system L amino acid transporters, induces apoptosis in cancer cells. Biol. Pharm. Bull. 2008, 31, 1096–1100. [Google Scholar] [CrossRef] [Green Version]
  87. Kobayashi, K.; Ohnishi, A.; Promsuk, J.; Shimizu, S.; Kanai, Y.; Shiokawa, Y.; Nagane, M. Enhanced tumor growth elcited by L-type amino acid transporter 1 in human malignant glioma cells. Neurosurgery 2008, 62, 493–504. [Google Scholar] [CrossRef]
  88. Avissar, N.E.; Sax, H.C.; Toia, L. In human entrocytes, GLN transport and ASCT2 surface expression induced by short-term EGF are MAPK, PI3K, and Rho-dependent. Dig. Dis. Sci. 2008, 53, 2113–2125. [Google Scholar] [CrossRef]
  89. Liu, P.; Ge, M.; Hu, J.; Li, X.; Che, L.; Sun, K.; Cheng, L.; Huang, Y.; Pilo, M.G.; Cigliano, A.; et al. A functional mammalian target of rapamycin complex 1 signaling is indispensable for c-Myc-driven hepatocarcinogenesis. Hepatology 2017, 66, 167–181. [Google Scholar] [CrossRef] [Green Version]
  90. Reynolds, M.R.; Lane, A.N.; Robertson, B.; Kemp, S.; Liu, Y.; Hill, B.G.; Dean, D.C.; Clem, B.F. Control of glutamine metabolism by the tumor suppressor Rb. Oncogene 2014, 33, 556–566. [Google Scholar] [CrossRef] [Green Version]
  91. Bröer, A.; Rahimi, F.; Bröer, S. Deletion of amino acid transporter ASCT2 (SLC1A5) reveals an essential role for transporters SNAT1 (SLC38A1) and SNAT2 (SLC38A2) to sustain glutaminolysis in cancer cells. J. Biol. Chem. 2016, 291, 13194–13205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Schioth, H.B.; Roshanbin, S.; Hagglund, M.G.; Fredriksson, R. Evolutionary origin of amino acid transporter families SLC32, SLC36 and SLC38 and physiological, pathological and therapeutic aspects. Mol. Asp. Med. 2013, 34, 571–585. [Google Scholar] [CrossRef] [PubMed]
  93. Cormerais, Y.; Giuliano, S.; LeFloch, R.; Front, B.; Durivault, J.; Tambutte, E.; Massard, P.A.; de la Ballina, L.R.; Endou, H.; Wempe, M.F.; et al. Genetic disruption of the multifunctional CD98/LAT1 complex demonstrates the key role of essential amino acid transport in the control of mTORC1 and tumor growth. Cancer Res. 2016, 76, 4481–4492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Milkereit, R.; Persaud, A.; Vanoaica, L.; Guetg, A.; Verrey, F.; Rotin, D. LAPTM4b recruits the LAT1-4F2hc Leu transporter to lysosomes and promotes mTORC1 activation. Nat. Commun. 2015, 6, 7250. [Google Scholar] [CrossRef] [Green Version]
  95. Imai, H.; Kaira, K.; Oriuchi, N.; Shimizu, K.; Tominaga, H.; Yanagitani, N.; Sunaga, N.; Ishizuka, T.; Nagamori, S.; Promchan, K.; et al. Inhibition of L-type amino acid transporter 1 has antitumor activity in non-small cell lung cancer. Anticancer Res. 2010, 30, 4819–4828. [Google Scholar]
  96. Shennan, D.B.; Thomson, J. Inhibition of system L (LAT1/CD98hc) reduces the growth of cultured human breast cancer cells. Oncol. Rep. 2008, 20, 885–889. [Google Scholar] [CrossRef] [Green Version]
  97. Ohkame, H.; Masuda, H.; Ishii, Y.; Kanai, Y. Expression of L-type amino acid transporter 1 (LAT1) and 4F2 heavy chain (4F2hc) in liver tumor lesions of rat models. J. Surg. Oncol. 2001, 78, 265–272. [Google Scholar] [CrossRef]
  98. Kim, C.S.; Moon, I.S.; Park, J.H.; Shin, W.C.; Chun, H.S.; Lee, S.Y.; Kook, J.K.; Kim, H.J.; Park, J.C.; Endou, H.; et al. Inhibition of L-type amino acid transporter modulates the expression of cell cycle regulatory factors in KB oralcancer cells. Biol. Pharm. Bull. 2010, 33, 1117–1121. [Google Scholar] [CrossRef] [Green Version]
  99. Sarita Rajender, P.; Ramasree, D.; Bhargavi, K.; Vasavi, M.; Uma, V. Selective inhibition of proteins regulating CDK/cyclin complexes: Strategy against cancer—A review. J. Recept. Signal Transduct. Res. 2010, 30, 206–213. [Google Scholar] [CrossRef]
  100. Wise, D.R.; DeBerardinis, R.J.; Mancuso, A.; Sayed, N.; Zhang, X.Y.; Pfeiffer, H.K.; Nissim, I.; Daikhin, E.; Yudkoff, M.; McMahon, S.B.; et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc. Natl. Acad. Sci. USA 2008, 105, 18782–18787. [Google Scholar] [CrossRef] [Green Version]
  101. Jeon, Y.J.; Khelifa, S.; Ratnikov, B.; Scott, D.A.; Feng, Y.; Parisi, F.; Ruller, C.; Lau, E.; Kim, H.; Brill, L.M.; et al. Regulation of Glutamine Carrier Proteins by RNF5 Determines Breast Cancer Response to ER Stress-Inducing Chemotherapies. Cancer Cell 2015, 27, 354–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Venmar, K.T.; Kimmel, D.W.; Cliffel, D.E.; Fingleton, B. IL4 receptor α mediates enhanced glucose and glutamine metabolism to support breast cancer growth. Biochim. Biophys. Acta Mol. Cell Res. 2015, 1853, 1219–1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Wellen, K.E.; Lu, C.; Mancuso, A.; Lemons, J.M.S.; Ryczko, M.; Dennis, J.W.; Rabinowitz, J.D.; Coller, H.A.; Thompson, C.B. The hexosamine biosynthetic pathway couples growth factor-induced glutamine uptake to glucose metabolism. Genes Dev. 2010, 24, 2784–2799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Ren, P.; Yue, M.; Xiao, D.; Xiu, R.; Gan, L.; Liu, H.; Qing, G. ATF4 and N-Myc coordinate glutamine metabolism in MYCN-amplified neuroblastoma cells through ASCT2 activation. J. Pathol. 2015, 235, 90–100. [Google Scholar] [CrossRef] [PubMed]
  105. Dong, J.; Xiao, D.; Zhao, Z.; Ren, P.; Li, C.; Hu, Y.; Shi, J.; Su, H.; Wang, L.; Liu, H.; et al. Epigenetic silencing of microRNA-137 enhances ASCT2 expression and tumor glutamine metabolism. Oncogenesis 2017, 6, e356. [Google Scholar] [CrossRef] [Green Version]
  106. Luo, M.; Wu, L.; Zhang, K.; Wang, H.; Zhang, T.; Gutierrez, L.; O’Connell, D.; Zhang, P.; Li, Y.; Gao, T.; et al. miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 2018, 25, 1457–1472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Rajasinghe, L.D.; Hutchings, M.; Gupta, S.V. Delta-tocotrienol modulates glutamine dependence by inhibiting ASCT2 and LAT1 transporters in non-small cell lung cancer (NSCLC) cells: A metabolomic approach. Metabolites 2019, 9, 50. [Google Scholar] [CrossRef] [Green Version]
  108. Sevigny, C.M.; Sengupta, S.; Luo, Z.; Liu, X.; Hu, R.; Zhang, Z.; Jin, L.; Pearce, D.; Demas, D.; Shajahan-Haq, A.N.; et al. SLCs contribute to endocrine resistance in breast cancer: Role of SLC7A5 (LAT1). BioRxiv 2019. [Google Scholar] [CrossRef]
  109. Sharma, A.; Boise, L.H.; Shanmugam, M. Cancer metabolism and the evasion of apoptotic cell death. Cancers 2019, 11, 1144. [Google Scholar] [CrossRef] [Green Version]
  110. Shalini, S.; Dorstyn, L.; Dawar, S.; Kumar, S. Old, new and emerging functions of caspases. Cell Death Differ. 2015, 22, 526–539. [Google Scholar] [CrossRef] [Green Version]
  111. Wang, Q.; Holst, J. L-type amino acid transport and cancer: Targeting the mTORC1 pathway to inhibit neoplasia. Am. J. Cancer Res. 2015, 5, 1281–1294. [Google Scholar] [PubMed]
  112. Betz, C.; Hall, M.N. Where is mTOR and what is it doing there? J. Cell Biol. 2013, 203, 563–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. van Geldermalsen, M.; Wang, Q.; Nagarajah, R.; Marshall, A.D.; Thoeng, A.; Gao, D.; Ritchie, W.; Feng, Y.; Bailey, C.G.; Deng, N.; et al. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene 2016, 35, 3201–3208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wang, Q.; Beaumont, K.A.; Otte, N.J.; Font, J.; Bailey, C.G.; van Geldermalsen, M.; Sharp, D.M.; Tiffen, J.C.; Ryan, R.M.; Jormakka, M.; et al. Targeting glutamine transport to suppress melanoma cell growth. Int. J. Cancer 2014, 135, 1060–1071. [Google Scholar] [CrossRef]
  115. Willems, L.; Jacque, N.; Jacquel, A.; Neveux, N.; Trovati Maciel, T.; Lambert, M.; Schmitt, A.; Poulain, L.; Green, A.S.; Uzunov, M.; et al. Inhibiting glutamine uptake represents an attractive new strategy for treating acute myeloid leukemia. Blood 2013, 122, 3521–3532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Ye, J.; Huang, Q.; Xu, J.; Huang, J.; Wang, J.; Zhong, W.; Chen, W.; Lin, X.; Lin, X. Targeting of glutamine transporter ASCT2 and glutamine synthetase suppresses gastric cancer cell growth. J. Cancer Res. Clin. Oncol. 2018, 144, 821–833. [Google Scholar] [CrossRef] [Green Version]
  117. Fan, X.; Ross, D.D.; Arakawa, H.; Ganapathy, V.; Tamai, I.; Nakanishi, T. Impact of system L amino acid transporter 1 (LAT1) on proliferation of human ovarian cancer cells: A possible target for combination therapy with anti-proliferative aminopeptidase inhibitors. Biochem. Pharmacol. 2010, 80, 811–818. [Google Scholar] [CrossRef]
  118. Nicklin, P.; Bergman, P.; Zhang, B.; Triantafellow, E.; Wang, H.; Nyfeler, B.; Yang, H.; Hild, M.; Kung, C.; Wilson, C.; et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 2009, 136, 521–534. [Google Scholar] [CrossRef] [Green Version]
  119. Cormerais, Y.; Massard, P.A.; Vucetic, M.; Giuliano, S.; Tambutté, E.; Durivault, J.; Vial, V.; Endou, H.; Wempe, M.F.; Parks, S.K.; et al. The glutamine transporter ASCT2 (SLC1A5) promotes tumor growth independently of the amino acid transporter LAT1 (SLC7A5). J. Biol. Chem. 2018, 293, 2877–2887. [Google Scholar] [CrossRef] [Green Version]
  120. Bothwell, P.J.; Kron, C.D.; Wittke, E.F.; Czerniak, B.N.; Bode, B.P. Targeted suppression and knockout of ASCT2 or LAT1 in epithelial and mesenchymal human liver cancer cells fail to inhibit growth. Int. J. Mol. Sci. 2018, 19, 2093. [Google Scholar] [CrossRef] [Green Version]
  121. Shi, L.; Luo, W.; Huang, W.; Huang, S.; Huang, G. Downregulation of L-type amino acid transporter 1 expression inhibits the growth, migration and invasion of gastric cancer cells. Oncol. Lett. 2013, 6, 106–112. [Google Scholar] [CrossRef] [Green Version]
  122. Ichinoe, M.; Yanagisawa, N.; Mikami, T.; Hana, K.; Nakada, N.; Endou, H.; Okayasu, I.; Murakumo, Y. L-Type amino acid transporter 1 (LAT1) expression in lymph node metastasis of gastric carcinoma: Its correlation with size of metastatic lesion and Ki-67 labeling. Pathol. Res. Pract. 2015, 211, 533–538. [Google Scholar] [CrossRef] [PubMed]
  123. Cluntun, A.A.; Lukey, M.J.; Cerione, R.A.; Locasale, J.W. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer 2017, 3, 169–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wise, D.R.; Thompson, C.B. Glutamine addiction: A new therapeutic target in cancer. Trends Biochem. Sci. 2010, 35, 427–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Scalise, M.; Pochini, L.; Galluccio, M.; Console, L.; Indiveri, C. Glutamine transport and mitochondrial metabolism in cancer cell growth. Front. Oncol. 2017, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Yang, L.; Venneti, S.; Nagrath, D. Glutaminolysis: A hallmark of cancer metabolism. Annu. Rev. Biomed. Eng. 2017, 19, 163–194. [Google Scholar] [CrossRef] [PubMed]
  127. Ben-Baruch, A. Host microenvironment in breast cancer development: Inflammatory cells, cytokines and chemokines in breast cancer progression: Reciprocal tumor–microenvironment interactions. Breast Cancer Res. 2002, 5, 31–36. [Google Scholar] [CrossRef] [Green Version]
  128. Akdis, M.; Burgler, S.; Crameri, R.; Eiwegger, T.; Fujita, H.; Gomez, E.; Klunker, S.; Meyer, N.; O’Mahony, L.; Palomares, O.; et al. Interleukins, from 1 to 37, and interferon-γ: Receptors, functions, and roles in diseases. J. Allergy Clin. Immunol. 2011, 127, 701–721. [Google Scholar] [CrossRef]
  129. Duan, Y.; Li, F.; Li, Y.; Tang, Y.; Kong, X.; Feng, Z.; Anthony, T.G.; Watford, M.; Hou, Y.; Wu, G.; et al. The role of leucine and its metabolites in protein and energy metabolism. Amino Acids 2016, 48, 41–51. [Google Scholar] [CrossRef]
  130. Wahi, K.; Holst, J. ASCT2: A potential cancer drug target. Expert Opin. Ther. Targets 2019, 23, 555–558. [Google Scholar] [CrossRef]
  131. Bhutia, Y.D.; Babu, E.; Ramachandran, S.; Ganapathy, V. Amino acid transporters in cancer and their relevance to “glutamine addiction”: Novel targets for the design of a new class of anticancer drugs. Cancer Res. 2015, 75, 1782–1788. [Google Scholar] [CrossRef] [Green Version]
  132. Jin, S.E.; Jin, H.E.; Hong, S.S. Targeting L-type amino acid transporter 1 for anticancer therapy: Clinical impact from diagnostics to therapeutics. Expert Opin. Ther. Targets 2015, 19, 1319–1337. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, C.; Xu, J.; Xue, S.; Ye, J. Prognostic value of L-type amino acid transporter 1 (LAT1) in various cancers: A meta-analysis. Mol. Diagn. Ther. 2020. [Google Scholar] [CrossRef]
  134. Lu, J.J.; Li, P.; Yang, Y.; Wang, L.; Zhang, Y.; Zhu, J.Y.; Zhu, X.R.; Chen, M.B. Prognostic value of LAT-1 status in solid cancer: A systematic review and meta-analysis. PLoS ONE 2020, 15, e0233629. [Google Scholar] [CrossRef] [PubMed]
  135. Kaira, K.; Kawashima, O.; Endoh, H.; Imaizumi, K.; Goto, Y.; Kamiyoshihara, M.; Sugano, M.; Yamamoto, R.; Osaki, T.; Tanaka, S.; et al. Expression of amino acid transporter (LAT1 and 4F2hc) in pulmonary pleomorphic carcinoma. Hum. Pathol. 2019, 84, 142–149. [Google Scholar] [CrossRef] [PubMed]
  136. Kaira, K.; Oriuchi, N.; Imai, H.; Shimizu, K.; Yanagitani, N.; Sunaga, N.; Hisada, T.; Ishizuka, T.; Kanai, Y.; Nakajima, T.; et al. Prognostic significance of L-type amino acid transporter 1 (LAT1) and 4F2 heavy chain (CD98) expression in stage I pulmonary adenocarcinoma. Lung Cancer 2009, 66, 120–126. [Google Scholar] [CrossRef]
  137. Ebara, T.; Kaira, K.; Saito, J.; Shioya, M.; Asao, T.; Takahashi, T.; Sakurai, H.; Kanai, Y.; Kuwano, H.; Nakano, T. L-type amino-acid transporter 1 expression predicts the response to preoperative hyperthermo-chemoradiotherapy for advanced rectal cancer. Anticancer Res. 2010, 30, 4223–4227. [Google Scholar] [CrossRef]
  138. Ogawa, H.; Kaira, K.; Motegi, Y.; Yokobori, T.; Takada, T.; Katoh, R.; Osone, K.; Takahashi, R.; Katayama, C.; Oyama, T.; et al. Role of amino acid transporter expression as a prognostic marker in patients with surgically resected colorectal cancer. Anticancer Res. 2019, 39, 2535–2543. [Google Scholar] [CrossRef]
  139. Sato, K.; Miyamoto, M.; Takano, M.; Furuya, K.; Tsuda, H. Significant relationship between the LAT1 expression pattern and chemoresistance in ovarian clear cell carcinoma. Virchows Arch. 2019, 474, 701–710. [Google Scholar] [CrossRef]
  140. Toyoda, M.; Kaira, K.; Ohshima, Y.; Ishioka, N.S.; Shino, M.; Sakakura, K.; Takayasu, Y.; Takahashi, K.; Tominaga, H.; Oriuchi, N.; et al. Prognostic significance of amino-acid transporter expression (LAT1, ASCT2, and xCT) in surgically resected tongue cancer. Br. J. Cancer 2014, 110, 2506–2513. [Google Scholar] [CrossRef] [Green Version]
  141. Isoda, A.; Kaira, K.; Iwashina, M.; Oriuchi, N.; Tominaga, H.; Nagamori, S.; Kanai, Y.; Oyama, T.; Asao, T.; Matsumoto, M.; et al. Expression of L-type amino acid transporter 1 (LAT1) as a prognostic and therapeutic indicator in multiple myeloma. Cancer Sci. 2014, 105, 1496–1502. [Google Scholar] [CrossRef] [PubMed]
  142. Shimizu, A.; Kaira, K.; Kato, M.; Yasuda, M.; Takahashi, A.; Tominaga, H.; Oriuchi, N.; Nagamori, S.; Kanai, Y.; Oyama, T.; et al. Prognostic significance of L-type amino acid transporter 1 (LAT1) expression in cutaneous melanoma. Melanoma Res. 2015, 25, 399–405. [Google Scholar] [CrossRef] [PubMed]
  143. Yothaisong, S.; Namwat, N.; Yongvanit, P.; Khuntikeo, N.; Puapairoj, A.; Jutabha, P.; Anzai, N.; Tassaneeyakul, W.; Tangsucharit, P.; Loilome, W. Increase in L-type amino acid transporter 1 expression during cholangiocarcinogenesis caused by liver fluke infection and its prognostic significance. Parasitol. Int. 2017, 66, 471–478. [Google Scholar] [CrossRef] [PubMed]
  144. Kaira, K.; Sunose, Y.; Ohshima, Y.; Ishioka, N.S.; Arakawa, K.; Ogawa, T.; Sunaga, N.; Shimizu, K.; Tominaga, H.; Oriuchi, N.; et al. Clinical significance of L-type amino acid transporter 1 expression as a prognostic marker and potential of new targeting therapy in biliary tract cancer. BMC Cancer 2013, 13, 482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Yanagisawa, N.; Hana, K.; Nakada, N.; Ichinoe, M.; Koizumi, W.; Endou, H.; Okayasu, I.; Murakumo, Y. High expression of L-type amino acid transporter 1 as a prognostic marker in bile duct adenocarcinomas. Cancer Med. 2014, 3, 1246–1255. [Google Scholar] [CrossRef] [PubMed]
  146. Nikkuni, O.; Kaira, K.; Toyoda, M.; Shino, M.; Sakakura, K.; Takahashi, K.; Tominaga, H.; Oriuchi, N.; Suzuki, M.; Iijima, M.; et al. Expression of amino acid transporters (LAT1 and ASCT2) in patients with stage III/IV laryngeal squamous cell carcinoma. Pathol. Oncol. Res. 2015, 21, 1175–1181. [Google Scholar] [CrossRef]
  147. Maimaiti, M.; Sakamoto, S.; Yamada, Y.; Sugiura, M.; Rii, J.; Takeuchi, N.; Imamura, Y.; Furihata, T.; Ando, K.; Higuchi, K.; et al. Expression of L-type amino acid transporter 1 as a molecular target for prognostic and therapeutic indicators in bladder carcinoma. Sci. Rep. 2020, 10, 1292. [Google Scholar] [CrossRef]
  148. Yanagisawa, N.; Satoh, T.; Hana, K.; Ichinoe, M.; Nakada, N.; Endou, H.; Okayasu, I.; Murakumo, Y. L-amino acid transporter 1 may be a prognostic marker for local progression of prostatic cancer under expectant management. Cancer Biomark. 2015, 15, 365–374. [Google Scholar] [CrossRef]
  149. Altan, B.; Kaira, K.; Watanabe, A.; Kubo, N.; Bao, P.; Dolgormaa, G.; Bilguun, E.O.; Araki, K.; Kanai, Y.; Yokobori, T.; et al. Relationship between LAT1 expression and resistance to chemotherapy in pancreatic ductal adenocarcinoma. Cancer Chemother. Pharmacol. 2018, 81, 141–153. [Google Scholar] [CrossRef]
  150. Maeda, S.; Nakazato, Y.; Hayashi, K.; Nishihira, M.; Inoue, T.; Araki, O.; Karube, Y.; Kobayashi, S.; Chida, M. L-Type Amino Acid Transporter 1 Immunoreactivity as a Possible Diagnostic and Prognostic Marker of Thymic Carcinoma. Tohoku J. Exp. Med. 2018, 246, 167–174. [Google Scholar] [CrossRef] [Green Version]
  151. Toda, K.; Nishikawa, G.; Iwamoto, M.; Itatani, Y.; Takahashi, R.; Sakai, Y.; Kawada, K. Clinical role of ASCT2 (SLC1A5) in KRAS-mutated colorectal cancer. Int. J. Mol. Sci. 2017, 18, 1632. [Google Scholar] [CrossRef] [Green Version]
  152. Bjersand, K.; Seidal, T.; Sundström-Poromaa, I.; Åkerud, H.; Skirnisdottir, I. The clinical and prognostic correlation of HRNPM and SLC1A5 in pathogenesis and prognosis in epithelial ovarian cancer. PLoS ONE 2017, 12, e0179363. [Google Scholar] [CrossRef]
  153. Bernhardt, S.; Bayerlová, M.; Vetter, M.; Wachter, A.; Mitra, D.; Hanf, V.; Lantzsch, T.; Uleer, C.; Peschel, S.; John, J.; et al. Proteomic profiling of breast cancer metabolism identifies SHMT2 and ASCT2 as prognostic factors. Breast Cancer Res. 2017, 19, 112. [Google Scholar] [CrossRef] [Green Version]
  154. Ge, N.J.; Shi, Z.Y.; Yu, X.H.; Huang, X.J.; Wu, Y.S.; Chen, Y.Y.; Zhang, J.; Yang, Y.F. Genetic Variants in ASCT2 Gene are Associated with the Prognosis of Transarterial Chemoembolisation-Treated Early- Stage Hepatocelluar Carcinoma. Asian Pac. J. Cancer Prev. 2015, 16, 4103–4107. [Google Scholar] [CrossRef]
  155. Yazawa, T.; Shimizu, K.; Kaira, K.; Nagashima, T.; Ohtaki, Y.; Atsumi, J.; Obayashi, K.; Nagamori, S.; Kanai, Y.; Oyama, T.; et al. Clinical significance of coexpression of L-type amino acid transporter 1 (LAT1) and ASC amino acid transporter 2 (ASCT2) in lung adenocarcinoma. Am. J. Transl. Res. 2015, 7, 1126–1139. [Google Scholar]
  156. Honjo, H.; Kaira, K.; Miyazaki, T.; Yokobori, T.; Kanai, Y.; Nagamori, S.; Oyama, T.; Asao, T.; Kuwano, H. Clinicopathological significance of LAT1 and ASCT2 in patients with surgically resected esophageal squamous cell carcinoma. J. Surg. Oncol. 2016, 113, 381–389. [Google Scholar] [CrossRef]
  157. Hafliger, P.; Charles, R.P. The L-type amino acid transporter LAT1-An Emerging Target in Cancer. Int. J. Mol. Sci. 2019, 20, 2428. [Google Scholar] [CrossRef] [Green Version]
  158. Hayashi, K.; Anzai, N. Novel therapeutic approaches targeting L-type amino acid transporters for cancer treatment. World J. Gastrointest. Oncol. 2017, 9, 21–29. [Google Scholar] [CrossRef]
  159. Jiang, H.; Zhang, N.; Tang, T.; Feng, F.; Sun, H.; Qu, W. Target the human Alanine/Serine/Cysteine Transporter 2(ASCT2): Achievement and future for novel cancer therapy. Pharmacol. Res. 2020, 158, 104844. [Google Scholar] [CrossRef]
  160. Grewer, C.; Grabsch, E. New inhibitors for the neutral amino acid transporter ASCT2 reveal its Na+-dependent anion leak. J. Physiol. 2004, 557, 747–759. [Google Scholar] [CrossRef]
  161. Esslinger, C.S.; Cybulski, K.A.; Rhoderick, J.F. Nγ-Aryl glutamine analogues as probes of the ASCT2 neutral amino acid transporter binding site. Bioorg. Med. Chem. 2005, 13, 1111–1118. [Google Scholar] [CrossRef]
  162. Ndaru, E.; Garibsingh, R.A.; Shi, Y.; Wallace, E.; Zakrepine, P.; Wang, J.; Schlessinger, A.; Grewer, C. Novel alanine serine cysteine transporter 2 (ASCT2) inhibitors based on sulfonamide and sulfonic acid ester scaffolds. J. Gen. Physiol. 2019, 151, 357–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Foster, A.C.; Rangel-Diaz, N.; Staubli, U.; Yang, J.Y.; Penjwini, M.; Viswanath, V.; Li, Y.X. Phenylglycine analogs are inhibitors of the neutral amino acid transporters ASCT1 and ASCT2 and enhance NMDA receptor-mediated LTP in rat visual cortex slices. Neuropharmacology 2017, 126, 70–83. [Google Scholar] [CrossRef]
  164. Colas, C.; Grewer, C.; Otte, N.J.; Gameiro, A.; Albers, T.; Singh, K.; Shere, H.; Bonomi, M.; Holst, J.; Schlessinger, A. Ligand discovery for the Alanine-Serine-Cysteine Transporter (ASCT2, SLC1A5) from homology modeling and virtual screening. PLoS Comput. Biol. 2015, 11, e1004477. [Google Scholar] [CrossRef] [Green Version]
  165. Singh, K.; Tanui, R.; Gameiro, A.; Eisenberg, G.; Colas, C.; Schlessinger, A.; Grewer, C. Structure activity relationships of benzylproline-derived inhibitors of the glutamine transporter ASCT2. Bioorg. Med. Chem. Lett. 2017, 27, 398–402. [Google Scholar] [CrossRef] [Green Version]
  166. Suzuki, M.; Toki, H.; Furuya, A.; Ando, H. Establishment of monoclonal antibodies against cell surface domains of ASCT2/SLC1A5 and their inhibition of glutamine-dependent tumor cell growth. Biochem. Biophys. Res. Commun. 2017, 482, 651–657. [Google Scholar] [CrossRef]
  167. Kasai, N.; Sasakawa, A.; Hosomi, K.; Poh, T.W.; Chua, B.L.; Yong, W.P.; So, J.; Chan, S.L.; Soong, R.; Kono, K.; et al. Anti-tumor efficacy evaluation of a novel monoclonal antibody targeting neutral amino acid transporter ASCT2 using patient-derived xenograft mouse models of gastric cancer. Am. J. Transl. Res. 2017, 9, 3399–3410. [Google Scholar]
  168. Chiu, M.; Sabino, C.; Taurino, G.; Bianchi, M.G.; Andreoli, R.; Giuliani, N.; Bussolati, O. GPNA inhibits the sodium-independent transport system l for neutral amino acids. Amino Acids 2017, 49, 1365–1372. [Google Scholar] [CrossRef]
  169. Ohshima, Y.; Kaira, K.; Yamaguchi, A.; Oriuchi, N.; Tominaga, H.; Nagamori, S.; Kanai, Y.; Yokobori, T.; Miyazaki, T.; Asao, T.; et al. Efficacy of system L amino acid transporter 1 inhibition as a therapeutic target in esophageal squamous cell carcinoma. Cancer Sci. 2016, 107, 1499–1505. [Google Scholar] [CrossRef]
  170. Choi, D.W.; Kim, D.K.; Kanai, Y.; Wempe, M.F.; Endou, H.; Kim, J.K. JPH203, a selective L-type amino acid transporter 1 inhibitor, induces mitochondria-dependent apoptosis in Saos2 human osteosarcoma cells. Korean J. Physiol. Pharmacol. 2017, 21, 599–607. [Google Scholar] [CrossRef] [Green Version]
  171. Rosilio, C.; Nebout, M.; Imbert, V.; Griessinger, E.; Neffati, Z.; Benadiba, J.; Hagenbeek, T.; Spits, H.; Reverso, J.; Ambrosetti, D.; et al. L-type amino-acid transporter 1 (LAT1): A therapeutic target supporting growth and survival of T-cell lymphoblastic lymphoma/T-cell acute lymphoblastic leukemia. Leukemia 2015, 29, 1253–1266. [Google Scholar] [CrossRef]
  172. Yun, D.W.; Lee, S.A.; Park, M.G.; Kim, J.S.; Yu, S.K.; Park, M.R.; Kim, S.G.; Oh, J.S.; Kim, C.S.; Kim, H.J.; et al. JPH203, an L-type amino acid transporter 1–selective compound, induces apoptosis of YD-38 human oral cancer cells. J. Pharmacol. Sci. 2014, 124, 208–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Muto, Y.; Furihata, T.; Kaneko, M.; Higuchi, K.; Okunushi, K.; Morio, H.; Reien, Y.; Uesato, M.; Matsubara, H.; Anzai, N. Different response profiles of gastrointestinal cancer cells to an L-type amino acid transporter inhibitor, JPH203. Anticancer Res. 2019, 39, 159–165. [Google Scholar] [CrossRef] [PubMed]
  174. Okunushi, K.; Furihata, T.; Morio, H.; Muto, Y.; Higuchi, K.; Kaneko, M.; Otsuka, Y.; Ohno, Y.; Watanabe, Y.; Reien, Y.; et al. JPH203, a newly developed anti-cancer drug, shows a preincubation inhibitory effect on L-type amino acid transporter 1 function. J. Pharmacol. Sci. 2020, 144, 16–22. [Google Scholar] [CrossRef] [PubMed]
  175. Geier, E.G.; Schlessinger, A.; Fan, H.; Gable, J.E.; Irwin, J.J.; Sali, A.; Giacomini, K.M. Structure-based ligand discovery for the Large-neutral Amino Acid Transporter 1, LAT-1. Proc. Natl. Acad. Sci. USA 2013, 110, 5480–5485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Kongpracha, P.; Nagamori, S.; Wiriyasermkul, P.; Tanaka, Y.; Kaneda, K.; Okuda, S.; Ohgaki, R.; Kanai, Y. Structure-activity relationship of a novel series of inhibitors for cancer type transporter L-type amino acid transporter 1 (LAT1). J. Pharmacol. Sci. 2017, 133, 96–102. [Google Scholar] [CrossRef] [PubMed]
  177. Napolitano, L.; Scalise, M.; Koyioni, M.; Koutentis, P.; Catto, M.; Eberini, I.; Parravicini, C.; Palazzolo, L.; Pisani, L.; Galluccio, M.; et al. Potent inhibitors of human LAT1 (SLC7A5) transporter based on dithiazole and dithiazine compounds for development of anticancer drugs. Biochem. Pharmacol. 2017, 143, 39–52. [Google Scholar] [CrossRef]
  178. Okano, N.; Naruge, D.; Kawai, K.; Kobayashi, T.; Nagashima, F.; Endou, H.; Furuse, J. First-in-human phase I study of JPH203, an L-type amino acid transporter 1 inhibitor, in patients with advanced solid tumors. Investig. New Drugs 2020. [Google Scholar] [CrossRef]
  179. Baniasadi, S.; Chairoungdua, A.; Iribe, Y.; Kanai, Y.; Endou, H.; Aisaki, K.-i.; Igarashi, K.; Kanno, J. Gene expression profiles in t24 human bladder carcinoma cells by inhibiting an l-type amino acid transporter, lat1. Arch. Pharm. Res. 2007, 30, 444–452. [Google Scholar] [CrossRef]
  180. Liang, Z.; Cho, H.T.; Williams, L.; Zhu, A.; Liang, K.; Huang, K.; Wu, H.; Jiang, C.; Hong, S.; Crowe, R.; et al. Potential biomarker of L-type amino acid transporter 1 in breast cancer progression. Nucl. Med. Mol. Imaging 2011, 45, 93–102. [Google Scholar] [CrossRef] [Green Version]
  181. Marshall, A.D.; van Geldermalsen, M.; Otte, N.J.; Anderson, L.A.; Lum, T.; Vellozzi, M.A.; Zhang, B.K.; Thoeng, A.; Wang, Q.; Rasko, J.E.J.; et al. LAT1 is a putative therapeutic target in endometrioid endometrial carcinoma. Int. J. Cancer 2016, 139, 2529–2539. [Google Scholar] [CrossRef] [PubMed]
  182. Yamauchi, K.; Sakurai, H.; Kimura, T.; Wiriyasermkul, P.; Nagamori, S.; Kanai, Y.; Kohno, N. System L amino acid transporter inhibitor enhances anti-tumor activity of cisplatin in a head and neck squamous cell carcinoma cell line. Cancer Lett. 2009, 276, 95–101. [Google Scholar] [CrossRef] [PubMed]
  183. Fukumoto, S.; Hanazono, K.; Fu, D.R.; Endo, Y.; Kadosawa, T.; Iwano, H.; Uchide, T. A new treatment for human malignant melanoma targeting L-type amino acid transporter 1 (LAT1): A pilot study in a canine model. Biochem. Biophys. Res. Commun. 2013, 439, 103–108. [Google Scholar] [CrossRef] [PubMed]
  184. Otsuki, H.; Kimura, T.; Yamaga, T.; Kosaka, T.; Suehiro, J.I.; Sakurai, H. Prostate cancer cells in different androgen receptor status employ different leucine transporters. Prostate 2017, 77, 222–233. [Google Scholar] [CrossRef] [PubMed]
  185. Wang, Q.; Bailey, C.G.; Ng, C.; Tiffen, J.; Thoeng, A.; Minhas, V.; Lehman, M.L.; Hendy, S.C.; Buchanan, G.; Nelson, C.C.; et al. Androgen receptor and nutrient signaling pathways coordinate the demand for increased amino acid transport during prostate cancer progression. Cancer Res. 2011, 71, 7525–7536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Yameen, B.; Choi, W.I.; Vilos, C.; Swami, A.; Shi, J.; Farokhzad, O.C. Insight into nanoparticle cellular uptake and intracellular targeting. J. Control. Release 2014, 190, 485–499. [Google Scholar] [CrossRef] [Green Version]
  187. Kou, L.; Bhutia, Y.D.; Yao, Q.; He, Z.; Sun, J.; Ganapathy, V. Transporter-guided delivery of nanoparticles to improve drug permeation across cellular barriers and drug exposure to selective cell types. Front. Pharmacol. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  188. Kharya, P.; Jain, A.; Gulbake, A.; Shilpi, S.; Jain, A.; Hurkat, P.; Majumdar, S.; Jain, S.K. Phenylalanine-coupled solid lipid nanoparticles for brain tumor targeting. J. Nanopart. Res. 2013, 15, 2022. [Google Scholar] [CrossRef]
  189. Li, L.; Di, X.; Wu, M.; Sun, Z.; Zhong, L.; Wang, Y.; Fu, Q.; Kan, Q.; Sun, J.; He, Z. Targeting tumor highly-expressed LAT1 transporter with amino acid-modified nanoparticles: Toward a novel active targeting strategy in breast cancer therapy. Nanomedicine 2017, 13, 987–998. [Google Scholar] [CrossRef]
  190. Ong, Z.Y.; Chen, S.; Nabavi, E.; Regoutz, A.; Payne, D.J.; Elson, D.S.; Dexter, D.T.; Dunlop, I.E.; Porter, A.E. Multibranched gold nanoparticles with intrinsic LAT-1 targeting capabilities for selective photothermal therapy of breast cancer. ACS Appl. Mater. Interfaces 2017, 9, 39259–39270. [Google Scholar] [CrossRef]
  191. Maekawa-Matsuura, M.; Fujieda, K.; Maekawa, Y.; Nishimura, T.; Nagase, K.; Kanazawa, H. LAT1-targeting thermoresponsive liposomes for effective cellular uptake by cancer cells. ACS Omega 2019, 4, 6443–6451. [Google Scholar] [CrossRef] [Green Version]
  192. Watabe, T.; Hatazawa, J. (18)F-FBPA as a tumor specific tracer of L-type amino acid transporter 1 (LAT1): PET evaluation in tumor and inflammation compared to (18)F-FDG and (11)C-methionine. Eur. J. Nucl. Med. Mol. Imaging 2015, 18, 321–331. [Google Scholar] [CrossRef]
  193. Nozaki, S.; Nakatani, Y.; Mawatari, A.; Shibata, N.; Hume, W.E.; Hayashinaka, E.; Wada, Y.; Doi, H.; Watanabe, Y. (18)F-FIMP: A LAT1-specific PET probe for discrimination between tumor tissue and inflammation. Sci. Rep. 2019, 9, 15718. [Google Scholar] [CrossRef] [PubMed]
  194. Verhoeven, J.; Baguet, T.; Piron, S.; Pauwelyn, G.; Bouckaert, C.; Descamps, B.; Raedt, R.; Vanhove, C.; De Vos, F.; Goethals, I. 2-[(18)F]FELP, a novel LAT1-specific PET tracer, for the discrimination between glioblastoma, radiation necrosis and inflammation. Nucl. Med. Biol. 2020, 82, 9–16. [Google Scholar] [CrossRef] [PubMed]
Figure 1. ASCT2 structure. (a) Bottom view highlighting the three protomers that constitute the ASCT2 homotrimer (PDB 6GCT); (b) Side view of the homotrimer (PDB 6GCT); (c) Membrane topology of a ASCT2 protomer with the scaffold domain in yellow and the transport domain in blue. The ECL2, that connects the two domains, is represented in red (adapted from [11,16] and created with BioRender.com). ECL: extracellular loop, HP: helical loop, TM: transmembrane segment.
Figure 1. ASCT2 structure. (a) Bottom view highlighting the three protomers that constitute the ASCT2 homotrimer (PDB 6GCT); (b) Side view of the homotrimer (PDB 6GCT); (c) Membrane topology of a ASCT2 protomer with the scaffold domain in yellow and the transport domain in blue. The ECL2, that connects the two domains, is represented in red (adapted from [11,16] and created with BioRender.com). ECL: extracellular loop, HP: helical loop, TM: transmembrane segment.
Cancers 13 00203 g001
Figure 2. LAT1 structure. (a) Representation of the LAT1-4F2hc complex, with 4F2hc in orange and LAT1 in blue (PDB 6IRT); (b) Membrane topology of LAT1 transporter with the 4F2hc in yellow and the functional light chain LAT1 in blue. The disulfide bridge (S-S), that connects the two chains, is represented in red (adapted from [25] and created with BioRender.com). H: short helix, TM: transmembrane domain.
Figure 2. LAT1 structure. (a) Representation of the LAT1-4F2hc complex, with 4F2hc in orange and LAT1 in blue (PDB 6IRT); (b) Membrane topology of LAT1 transporter with the 4F2hc in yellow and the functional light chain LAT1 in blue. The disulfide bridge (S-S), that connects the two chains, is represented in red (adapted from [25] and created with BioRender.com). H: short helix, TM: transmembrane domain.
Cancers 13 00203 g002
Figure 3. ASCT2, LAT1, and the hallmarks of cancer. Both ASCT2 and LAT1 have been associated with avoiding immune destruction [7,67,68,69,70,71,72], invasion and metastasis [47,73,74,75], angiogenesis [63,74,76,77,78], resisting cell death [20,29,79,80,81,82,83,84,85,86,87], proliferative signaling [43,52,87,88,89,90,91,92,93,94,95,96,97,98,99], and cellular energetics [90,100,101,102,103,104,105,106,107,108]. The activation/overexpression (green) or inactivation/subexpression (red) of those molecules consequently results in the dysregulation of a variety of key players on those hallmarks of cancer, culminating in cancer. The role of ASCT2 and LAT1 in genomic instability and mutation, inflammation, replicative immortality, and evading growth suppressors remains to be explored (created with BioRender.com).
Figure 3. ASCT2, LAT1, and the hallmarks of cancer. Both ASCT2 and LAT1 have been associated with avoiding immune destruction [7,67,68,69,70,71,72], invasion and metastasis [47,73,74,75], angiogenesis [63,74,76,77,78], resisting cell death [20,29,79,80,81,82,83,84,85,86,87], proliferative signaling [43,52,87,88,89,90,91,92,93,94,95,96,97,98,99], and cellular energetics [90,100,101,102,103,104,105,106,107,108]. The activation/overexpression (green) or inactivation/subexpression (red) of those molecules consequently results in the dysregulation of a variety of key players on those hallmarks of cancer, culminating in cancer. The role of ASCT2 and LAT1 in genomic instability and mutation, inflammation, replicative immortality, and evading growth suppressors remains to be explored (created with BioRender.com).
Cancers 13 00203 g003
Figure 4. Pathways affected by ASCT2 and LAT1. ASCT2 is responsible for the transport of neutral amino acids into the cell, namely glutamine (Gln), coupled with the transport of one neutral amino acid (AA) to the extracellular environment. Gln can be used to synthesize glutathione (GSH), an antioxidant which prevents oxidative stress. In turn, together with ASCT2-related downregulation of caspases-2 and -9 and LAT1-related downregulation of caspases-3 and -7, oxidative stress inhibits apoptosis. On the other hand, Gln can also be used to biosynthesize other elements that play key roles in cell growth or be transported to the mitochondria and enter the glutaminolysis process, yielding metabolites, such as α-KG, pyruvate and lactate, and generating NADH and FADH2, essential for ATP production. Similar to ASCT2, LAT1 is an amino acid exchanger, transporting one amino acid into the cytosol, like leucine (Leu), and another amino acid to the extracellular milieu, namely Gln. Leu is required for the activation of the mammalian target of rapamycin complex (mTORC1), which in turn activates the downstream effectors ribosomal protein S6 kinase 1 (S6K1) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) through phosphorylation. Those proteins play important roles in mRNA and protein biosynthesis, resulting in cell growth. In the nucleus, proto-oncogenes and tumor suppressors, such as c-Myc and E2F transcription factor 3 (E2F3), respectively, have been found to bind to promoter elements of the gene encoding ASCT2, resulting in altered Gln metabolism through SLC1A5 dysregulation (created with BioRender.com).
Figure 4. Pathways affected by ASCT2 and LAT1. ASCT2 is responsible for the transport of neutral amino acids into the cell, namely glutamine (Gln), coupled with the transport of one neutral amino acid (AA) to the extracellular environment. Gln can be used to synthesize glutathione (GSH), an antioxidant which prevents oxidative stress. In turn, together with ASCT2-related downregulation of caspases-2 and -9 and LAT1-related downregulation of caspases-3 and -7, oxidative stress inhibits apoptosis. On the other hand, Gln can also be used to biosynthesize other elements that play key roles in cell growth or be transported to the mitochondria and enter the glutaminolysis process, yielding metabolites, such as α-KG, pyruvate and lactate, and generating NADH and FADH2, essential for ATP production. Similar to ASCT2, LAT1 is an amino acid exchanger, transporting one amino acid into the cytosol, like leucine (Leu), and another amino acid to the extracellular milieu, namely Gln. Leu is required for the activation of the mammalian target of rapamycin complex (mTORC1), which in turn activates the downstream effectors ribosomal protein S6 kinase 1 (S6K1) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) through phosphorylation. Those proteins play important roles in mRNA and protein biosynthesis, resulting in cell growth. In the nucleus, proto-oncogenes and tumor suppressors, such as c-Myc and E2F transcription factor 3 (E2F3), respectively, have been found to bind to promoter elements of the gene encoding ASCT2, resulting in altered Gln metabolism through SLC1A5 dysregulation (created with BioRender.com).
Cancers 13 00203 g004
Figure 5. Clinical significance of ASCT2 and LAT1. LAT1 has been used in the clinic for diagnostic purposes due to its ability to transport PET probes and enhance nanoparticles delivery. Moreover, high LAT1 expression has been associated with chemoresistance in some types of cancer. ASCT2 and LAT1 have also been identified as therapeutic targets, with some studies reporting the successful synthesis of potential competitive or irreversible inhibitors of these transporters, as well as the use of monoclonal antibodies.
Figure 5. Clinical significance of ASCT2 and LAT1. LAT1 has been used in the clinic for diagnostic purposes due to its ability to transport PET probes and enhance nanoparticles delivery. Moreover, high LAT1 expression has been associated with chemoresistance in some types of cancer. ASCT2 and LAT1 have also been identified as therapeutic targets, with some studies reporting the successful synthesis of potential competitive or irreversible inhibitors of these transporters, as well as the use of monoclonal antibodies.
Cancers 13 00203 g005
Table 1. ASCT2 and LAT1 expression in cancer.
Table 1. ASCT2 and LAT1 expression in cancer.
Authors 1 (Year) [Ref]Cancer ModelMethodTransporter Expression in Cancer (p Value)
Liu et al. (2015) [28] Clear cell renal cell carcinomaRT-qPCRASCT2 upregulation (p = 0.007)
Huang et al. (2014) [29]Colorectal cancerIHCASCT2 upregulation (p < 0.001)
WB(p < 0.05)
Schulte et al. (2017) [30]Primary and metastatic colorectal cancerIHCASCT2 upregulation in cancer (p < 0.001) and in metastases (p < 0.001) compared to normal tissue
Marshall et al. (2017) [31]Endometrial carcinomaRisinger et al. [32] cohort analysis (Multiplex-qPCR)ASCT2 upregulation in serous (p < 0.001) and endometrioid (p < 0.001) subtypes compared to normal tissue
Lu et al. (2017) [33]Gastric cancerIHCASCT2 upregulation (p < 0.0001)
RT-qPCR(n = 32, p = 0.0046; n = 45, p < 0.0001; n = 80, p = 0.0147)
Sun et al. (2016) [34]Hepatocellular carcinomaIHCASCT2 upregulation (p = 0.009)
Shimizu et al. (2014) [35]Non-small cell lung cancerIHCHigher ASCT2 expression in non-AC compared to AC (p = 0.019)
Guo et al. (2018) [36]Epithelial ovarian cancerRT-qPCR and WBASCT2 upregulation (p < 0.001)
Kaira et al. (2015) [37]Pancreatic ductal carcinomaIHCASCT2 upregulation (p < 0.001)
Wang et al. (2015) [38]Prostate cancerTCGA cohort analysisASCT2 upregulation (p = 0.025) and higher ASCT2 expression in recurrent cancer compared to patients undergoing NHT (p < 0.001)
Namikawa et al. (2015) [39]Hepatocellular carcinomaIHCHigher ASCT2 expression compared to LAT1 (p < 0.001)
Kaira et al. (2015) [40]Ovarian cancerIHCHigher ASCT2 expression compared to LAT1 (p = 0.032), although no difference was found in epidermal ovarian cancer
Papin-Michault et al. (2016) [41]Brain metastasisIHCLAT1 upregulation (p < 0.001)
Betsunoh et al. (2013) [42]Clear cell renal cell carcinomaRT-qPCRLAT1 upregulation (p < 0.0001)
Hayase et al. (2017) [43]Colorectal cancer/Colonic adenomaIHCLAT1 upregulation compared to non-malignant tissue (NA) and colonic adenoma (p < 0.001)
Sakata et al. (2020) [44]Colorectal ACWBLAT1 upregulation compared to normal tissue (NA), with an increase in a sporadic adenoma-adenoma-carcinoma series (p < 0.0001)
IHC
Watanabe et al. (2014) [45]Endometrial endometrioid ACIHCLAT1 upregulation compared to atrophic endometrium (p < 0.01)
Sato et al. (2020) [46]Endometrial carcinomaIHCLAT1 upregulation in non-endometrioid compared to endometrioid carcinomas (p < 0.001)
Wang et al. (2016) [47]Gastric cancerRT-qPCRLAT1 upregulation (p < 0.01)
Li et al. (2013) [48]Hepatocellular carcinomaRT-qPCRLAT1 upregulation (p < 0.001)
Kaira et al. (2008) [49]Resectable stage I-III NSCLCIHCLAT1 upregulation (NA), higher LAT1 expression in SCC than AC (p < 0.001) and in non-AC than AC (p < 0.001)
Imai et al. (2009) [50]Stage I NSCLC IHCHigher LAT1 expression in SCC than AC (p < 0.001) and in non-AC than AC (p < 0.001)
Takeuchi et al. (2010) [51]NSCLCRT-PCRLAT1 upregulation (p < 0.05)
Kaira et al. (2011) [52]Resected NSCLCIHCHigher LAT1 expression in non-AC than in AC (p < 0.001)
Kaira et al. (2011) [53]NSCLC (recurrence after platinum-based chemotherapy)IHCHigher LAT1 expression in non-AC than in AC (p = 0.0022)
Kaira et al. (2012) [54]Pancreatic cancerIHCLAT1 upregulation (p < 0.001)
Yanagisawa et al. (2012) [55]Pancreatic ACIHCHigher LAT1 expression in IPMC compared to PDAC (p < 0.0001)
AC: adenocarcinoma, ASCT2: alanine, serine, cysteine transporter 2, IHC: immunohistochemistry, IPMC: intraductal papillary mucinous carcinoma, LAT-1: L-type amino acid transporter 1, NHT: neoadjuvant hormone therapy, NSCLC: non-small cell lung cancer, PDAC: pancreatic ductal adenocarcinoma, RT-qPCR: reverse transcription real-time polymerase chain reaction, SCC: squamous cell carcinoma, TCGA: The Cancer Genome Atlas, WB: western blot. 1 Published studies with no statistical significant results were not included [27,56,57,58,59,60,61,62,63,64,65].
Table 2. ASCT2 and LAT1 pharmacological inhibitors.
Table 2. ASCT2 and LAT1 pharmacological inhibitors.
TransporterInhibitorModelReference
ASCT2BenzylcysteineNormal kidney[160]
Benzylproline derivativesNormal kidney[165]
BenzylserineNormal kidney[160]
δTNSCLC[107]
GPNARat glioma[161]
MAb KM8094Gastric cancer[56]
Gastric cancer patient-derived xenograft mice[167]
MAb KM4008Colorectal cancer[166]
MAb KM4012Colorectal cancer[166]
MAb KM4018Colorectal cancer[166]
Phenylglycine analoguesWistar rat[163]
Normal kidney[163]
Sulfonamide/sulfonic acid ester scaffolds-based moleculesNormal kidney[162]
V-9302Normal kidney[83]
Breast cancer[83]
Colorectal cancer[83]
Lung cancer[83]
LAT1BCHBladder cancer [179]
Breast cancer[180]
Cervical cancer[86]
Cholangiocarcinoma[144]
Endometrial cancer[181]
Esophageal cancer[169]
Glioma[87]
Head and neck[182]
Melanoma[183]
NSCLC[95]
Osteosarcoma[86]
Ovarian cancer[58]
Prostate cancer[184,185]
Rat glioma[86]
δTNSCLC[107]
GPNABrain glioma[168]
Breast cancer[168]
Cervical cancer[168]
Hepatocellular carcinoma[168]
NSCLC[168]
JHP203Colorectal cancer[173,174]
Gastric cancer[173]
Head and neck[172]
Leukemia[171]
Osteosarcoma[170]
SKN103Cervical cancer[176]
NSCLC[176]
Pancreatic cancer[176]
δT: delta-tocotrienol, ASCT2: alanine, serine, cysteine transporter 2, BCH: 2-aminobicyclo[2,2,1]-heptane-2-carboxylic acid, GPNA: L-γ-glutamyl-p-nitroanilide, LAT-1: L-type amino acid transporter 1, MAb: monoclonal antibody, NSCLC: non-small cell lung cancer.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lopes, C.; Pereira, C.; Medeiros, R. ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation. Cancers 2021, 13, 203. https://doi.org/10.3390/cancers13020203

AMA Style

Lopes C, Pereira C, Medeiros R. ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation. Cancers. 2021; 13(2):203. https://doi.org/10.3390/cancers13020203

Chicago/Turabian Style

Lopes, Catarina, Carina Pereira, and Rui Medeiros. 2021. "ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation" Cancers 13, no. 2: 203. https://doi.org/10.3390/cancers13020203

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop